Androgen Receptor Nuclear Translocation Is Facilitated by the f-Actin Cross-Linking Protein Filamin
Daniel M. Ozanne,
Mark E. Brady,
Susan Cook,
Luke Gaughan,
David E. Neal and
Craig N. Robson
Prostate Research Group School of Surgical and Reproductive
Sciences Medical School, University of Newcastle upon Tyne
Newcastle upon Tyne, England NE2 4HH
 |
ABSTRACT
|
---|
The human androgen receptor (hAR) is a
ligand-dependent transcription factor responsible for the development
of the male phenotype. The mechanism whereby nuclear translocation of
the hAR is induced by its natural ligand 5
-dihydrotestosterone is a
phenomenon not fully understood. The two-hybrid interaction trap assay
has been used to isolate proteins that interact with the hAR in an
attempt to identify molecules involved in hAR transactivation and
movement. We have identified the actin-binding protein filamin, a
280-kDa component of the cytoskeleton, as an hAR interacting protein.
This interaction is ligand independent but is enhanced in its presence.
The functional significance of this interaction was analyzed using a
cell line deficient in filamin via transient expression of a green
fluorescent protein-hAR chimera. In filamin-deficient cells this
revealed that hAR remained cytoplasmic even after prolonged exposure to
synthetic ligand. Nuclear shuttling was restored when this cell line
regained wild-type expression of filamin. These data suggest a novel
role for filamin, implicating it as an important molecule in AR
movement from the cytoplasm to the nucleus.
 |
INTRODUCTION
|
---|
Steroid hormone receptors (SHRs) exact their effects after
activation by their cognate ligands and binding to specific responsive
elements on their target genes within the nucleus (reviewed in Refs. 1, 2). The cellular localization of different SHRs is varied and
dependent upon the specific receptor. Both the unliganded progesterone
receptor and estrogen receptor are nuclear in localization, whereas
unliganded glucocorticoid receptor (GR) exists within the cytoplasm and
is translocated to the nucleus upon activation (3, 4, 5). The
mineralocorticoid receptor is found both in the cytoplasm and the
nucleus in the inactivated form (6). The human androgen receptor (hAR)
presents a more complicated cellular distribution. Immunohistochemical
studies observing the endogenous hAR, and overexpression of hAR by
transient transfection, provide conflicting data on receptor
distribution in specific cell types (7).
The receptor becomes activated on binding of its cognate ligand,
5
-dihydrotestosterone (DHT), generated from testosterone by membrane
bound 5
-reductase. The hAR is a cis-acting transcription
factor essential for the growth and differentiation of cells within the
prostate and male external genitalia. It is also a pivotal molecule in
the development and progression of prostatic carcinoma.
Little is known about the mechanism of cytoplasmic translocation or
associated molecules that coordinate movement of the activated hAR to
the nucleus. Work with the hAR fused to a green fluorescent protein
(GFP) reporter has allowed the study of the movement of the hAR
in vivo. In Cos-7 cells, unliganded hAR is cytoplasmic and
is fully translocated to the nucleus within 30 min of steroid addition
(8). Although this study highlighted specific hAR kinetics, it did not
reveal a molecular mechanism of receptor movement. Specific hAR
kinetics has been linked to a bipartite nuclear targeting signal
between the DNA binding domain and the hinge region of the hAR. A
region of the hAR consisting of two clusters of basic amino acids,
separated by 10 residues, is necessary for full receptor nuclear
import, and is modulated by elements within the
NH2 and carboxyl-terminal regions (9).
The role of the cytoskeleton in the trafficking of steroid hormone
receptors has presented conflicting data. Studies involving a
progesterone receptor mutant with an inactive karyophilic signal
revealed that chemical disruption of the microtubule and actin network
neither prevented nor delayed the hormone-dependent transfer of the PR
mutant to the nucleus (10). Conversely, receptor trafficking studies
involving the GR fused to a GFP highlighted the cytoskeletal network as
an important structure in GR movement (11). Chemical disruption of the
cytoskeleton by colcemid blocks the okadaic acid-dependent inhibition
of hormone-dependent GR recycling to the nucleus. This implies that
elements within the cytoskeleton are required for GR movement and that
when the cytoskeleton is disrupted the normal shuttling of GR
via cytoskeletal tracts utilizing cytoskeletal associating motor
proteins is abrogated. A key molecule in this process is the cellular
chaperone heat shock protein 90 (Hsp90) that forms a stable
heterocomplex with the GR and has been shown to translocate to the
nucleus with the receptor (12). The role of Hsp90 in this interaction
may be to facilitate translocation by tethering the GR heterocomplex
with the cytoplasmic movement machinery. Addition of okadaic acid may
prevent the heterocomplex binding to these elements, allowing it to
move to the nucleus via diffusion.
GFP-visualized movement of SHRs has also been performed previously for
other members of the SHR superfamily (13, 14, 15, 16, 17). These dynamic studies
have detailed the subcellular distribution of SHRs both before and
after activation by ligand.
We have performed a yeast two-hybrid interaction trap assay (18) to
isolate proteins that interact specifically with the hAR. The aim of
this study was to identify novel hAR interacting proteins involved in
either movement or signaling from the cytoplasm to the nucleus. A cDNA
clone encoding a central portion of the cytoskeletal actin-binding
protein filamin (ABP 280) was isolated as an hAR interacting protein.
Using a GFP-hAR chimera, we have shown that hAR nuclear translocation
is abolished in a cell line deficient in the cytoskeletal protein
filamin, and hence that filamin appears to have a significant role to
play in nuclear translocation of the hAR.
 |
RESULTS
|
---|
The Actin-Binding Protein Filamin Is an hAR Interactor
The actin-binding protein filamin (ABP 280) was identified as
interacting with the human hAR in a two-hybrid interaction trap assay
using an hAR fragment comprising DNA-binding and ligand-binding domains
as bait. The clone isolated in the screen spanned an area of 334 amino
acids from amino acids 1,788 to 2,121, designated
Fil1788-2121. This
interaction is not dependent on ligand being present but is enhanced in
its presence (Fig. 1
). A separate
two-hybrid assay (data not shown) also identified
-filamin (19) as
an hAR-interacting protein. Both interacting clones span an area of
high homology between the two isoforms overlapping highly conserved
IgG-like repetitive regions. Androgen receptor constructs comprising
either the DNA-binding domain or the ligand-binding domain produced
only basal levels of ß-galactosidase activity. This suggests that the
region of the AR involved with filamin interaction possibly requires
the three-dimensional (3-D) structure created via the juxtaposition of
the two adjacent domains.

View larger version (24K):
[in this window]
[in a new window]
|
Figure 1. Relative ß-Galactosidase Activities of
Fil1788-2121 Cotransformed
with hAR Deletions as Indicated, in a Yeast Two-Hybrid LacZ Assay
The hAR deletion construct comprising residues 559918 presented the
highest activity in the presence of DHT. A reduced interaction was
observed with this construct in the absence of ligand. Other hAR
constructs used in the assay did not present a significant interaction
above that observed with empty vectors only. Bar chart shows
the average activity observed from two independent experiments
performed in triplicate.
|
|
Filamin and hAR Interact in Vitro
The interaction between filamin and hAR was analyzed using
in vitro transcription and translation of the proteins
labeled with 35S-methionine. Figure 2A
represents in vitro
transcribed/translated protein products before immunoprecipitation. A
minimal domain of interaction was sought using hAR truncations
corresponding to functional domains of the hAR. Figure 2B
demonstrates
that the filamin clone
Fil1788-2121 interacts
with the full-length hAR (amino acids 1918), the DNA-binding domain
and the steroid- binding domain (amino acids 559918), and the
steroid-binding domain alone (amino acids 624918). No interaction was
observed between
Fil1788-2121 and the
transactivation domain and the DNA-binding domain (amino acids 1624).
To further minimize this domain of interaction, hAR truncations
encompassing the steroid-binding domain including the hinge region of
the hAR were assessed for their ability to coprecipitate. Figure 2C
shows that all the constructs containing the hinge region of the hAR
coprecipitate with
Fil1788-2121 including the
hinge region of the hAR alone (amino acids 616674).

View larger version (62K):
[in this window]
[in a new window]
|
Figure 2. In Vitro Interaction of
Fil1788-2121 and hAR
A, Fil1788-2121, full-length hAR
(AR1-918), and three hAR deletion constructs
(hAR559-918, hAR1-624, hAR624-918)
were transcribed and translated in vitro with
35S-methionine. Radiolabeled protein products were mixed
and immunoprecipitated in the presence of DHT using an antibody
directed against a penta-His motif on the filamin clone. B,
Fil1788-2121 was observed to interact with the
full-length hAR (hAR1-918) and constructs
encoding the DNA-binding and steroid-binding domains
(hAR559-918) and the steroid-binding domain
alone (hAR624-918). No interaction was
observed between Fil1788-2121 and the
construct encoding the transactivation and DNA-binding domains of the
hAR (hAR1-624). C, hAR constructs encoding N-
and C-terminal deletions of hAR559-918 were
used to further define the area of interaction with
Fil1788-2121. hAR fragments containing the
hinge region of the hAR (residues 616674) showed an interaction with
Fil1788-2121. Large arrows
correspond to labeled Fil1788-2121.
|
|
The results obtained in two-hybrid interaction assay for the two
proteins indicated that both the DNA-binding and the steroid-binding
domains of the hAR were necessary for the filamin interaction. However,
these constructs are fused to either the GAL4 activation or
GAL4-binding domains, which may interfere with the correct folding of
native proteins and thus alter potential sites of interaction. When the
proteins are expressed in vitro, the interacting domains are
more closely related, in terms of folding, to the in vivo
state.
Filamin and hAR Interact in Vivo
Immunoprecipitation and subsequent Western analysis using hAR and
filamin antibodies (Fig. 3
) confirmed
this interaction. A mouse monoclonal antibody was used to
immunoprecipitate filamin protein from prostate LNCaP cells before
detection of hAR by Western analysis. A 98-kDa band, corresponding to
the hAR, was observed (lane 1), which was absent in control lanes
omitting extract or antibody (lanes 2 and 3, respectively). The normal
distribution of filamin has been well documented using
immunohistochemistry, occurring predominantly along stress fibers and
in the cell periphery (20). It is therefore likely that filamin-AR
interaction occurs primarily within the cytoplasm.

View larger version (37K):
[in this window]
[in a new window]
|
Figure 3. Filamin and hAR Interact in Vivo
Protein extract from LNCaP cells was used to determine whether the hAR
and filamin coprecipitate in vivo. Antifilamin antibody
was used to immunoprecipitate filamin and associating proteins.
Resulting protein complexes were resolved on a polyacrylamide gel and
Western analysis was performed with an anti-hAR antibody as depicted
above. Lane 1 demonstrates that the hAR associates with filamin in
LNCaP cells. No coimmunoprecipitating hAR protein was observed in lane
2 (antibody in the absence of cell extract) or lane 3 (cell extract in
the absence of antibody).
|
|
The Minimal Region of Interaction Spans a 150-Amino
Acid Fragment of Filamin
The minimal domain of filamin required to interact with hAR was
investigated by deleting residues from both the N- and C terminus of
the Fil1788-2121 cDNA
clone (Fig. 4A
). Deletion constructs in
pACT2 were tested for their ability to interact with hAR in yeast. The
ß-galactosidase activity of these clones was assessed in comparison
to the original filamin clone in pACT2. The relative ß-galactosidase
activities of these clones were quantified in a liquid LacZ assay (Fig. 4B
). The minimal domain of interaction was shown to lie within a
150-amino acid fragment encompassing repeats 18 and 19 of the filamin
IgG-like repeats. This area of interaction is similar to that
identified for the glycoprotein Ib
, which was found to interact
between repeats 17 and 20. The majority of the other proteins
identified as interactors of filamin appear to be focused around this
C-terminal portion of the protein, indicating that elements within
these IgG-like repeats are responsible for the specificity of
interaction with these varied cellular molecules.

View larger version (23K):
[in this window]
[in a new window]
|
Figure 4. Minimal Domain Mapping of the Interaction of hAR on
Filamin
Filamin deletion constructs were cotransformed into yeast strain
PJ694A with hAR559-918 in pAS21. Resultant
leu+, trp+ colonies were assayed for their ß-galactosidase activity.
The relative ß-galactosidase activity compared with
hAR559-918 was noted, as were the
cotransformants ability to grow on his-, leu-, trp- media in the
presence of 5 mM 3-AT, "+" denoting a comparable
activity. A 400-bp filamin construct was observed to have
ß-galactosidase activity comparable to that observed for the
Fil1788-2121 construct. The
graph represents the relative activities of the Filamin
deletions compared with the original bait construct,
Fil1788-2121. Data represents the average of
three independent experiments performed in triplicate. Error
bars represent the SD of three experiments.
|
|
Mutant Filamin Inhibits hAR-Dependent Transactivation
A transient transfection reporter gene assay was performed to
investigate the influence of Fil
1788-2121 on the
transactivation activity of hAR (Fig. 5A
). COS-7 cells were transfected with
plasmids encoding wild-type hAR (50 ng) and
Fil1788-2121 (0800 ng),
together with a luciferase reporter plasmid containing an enhancer
element comprising three androgen responsive elements (AREs), p
(ARE)3-Luc. As shown in Fig. 5
, hAR is activated
by the synthetic androgen ligand, mibolerone. Increasing amounts of
Fil1788-2121, in the presence or absence of androgen,
cause a dramatic reduction in the level of hAR transactivation.
Parallel experiments, using wild-type filamin in place of
Fil1788-2121, only caused a small reduction in the
mibolerone-induced AR activation, observed at the highest doses of
filamin (data not shown). A negative control promoter (1,659 bp)
derived from the matrix metalloproteinase 2 (MMP2) promoter, which
lacks defined SHR-responsive DNA elements and is known to be
unresponsive to androgens (our unpublished data) was used to
demonstrate that
Fil1788-2121 had no effect
on the inhibition of transactivation via a nonspecific promoter element
(Fig. 5B
). These results suggest that the truncated form of filamin,
Fil1788-2121, is acting as
a dominant negative protein by binding hAR and preventing activation of
target genes.

View larger version (26K):
[in this window]
[in a new window]
|
Figure 5. Fil1788-2121 Inhibits
hAR-Dependent Transactivation of a Target Gene but Has No Effect on a
Non-AR-Responsive Promoter Element
COS-7 cells were transfected with wild-type hAR and increasing
amounts of pCMV-Fil1788-2121 together with
p(ARE)3-Luc or p(MMP2 RE)3-Luc. In the absence
of Fil1788-2121, the hAR is activated by the
presence of 10 nM mibolerone. The addition of
increasing amounts of Fil1788-2121 results in
a rapid decrease in the transactivational activity of the hAR (A). A
non-AR-responsive MMP2 promoter is unaffected by increasing addition of
Fil1788-2121 (B).. Results shown
are the average of three independent experiments performed in
triplicate. Error bars represent the SD
of three experiments.
|
|
The ability of filamin to inhibit the activation potential of a target
gene has been reported previously (21). Tumor necrosis factor
receptor-associated factor (TRAF) 2 and tumor necrosis
factor-
activation of nuclear factor-
B and TRAF2
activation of the JNK/SAPK (c-jun N-terminal
kinase/stress-activated protein kinase) pathway is inhibited by
the addition of increasing amounts of filamin added in a similar
reporter gene assay. Interestingly, the filamin clone used encompassed
amino acids 16442118 of the full cDNA sequence, a fragment
overlapping the one identified in our study as an hAR interactor. It
would be tempting to speculate that the mechanism of filamin-induced
inhibition of target gene expression is similar in both systems.
A GFP-hAR Chimera Remains Cytoplasmic in a Filamin-Negative Cell
Line
To assess the functional significance of the filamin-hAR
interaction, the movement of the hAR in response to ligand was analyzed
in a cell line deficient in filamin. Previous reports regarding the GR
highlighted the importance of the cytoskeleton in the recycling of the
GR to the nucleus. A GFP-hAR fusion was used to visualize hAR
localization. An hAR construct was generated which lacked the
N-terminal transactivation domain
(GFP-hAR559-918),
previously shown to be constituitively nuclear in the absence of ligand
(22), to act as a positive control. A recent investigation with an
AR-GFP chimera reported that unliganded AR was cytoplasmic but rapidly
translocated to the nucleus upon addition of ligand (8). In the
filamin-deficient parental cell line (M2FIL-) we
observed the hAR to be cytoplasmic in location and remained cytoplasmic
even after prolonged exposure to synthetic ligand (Fig. 6
, A and B). However, in a stably
transfected derivative of this cell line expressing filamin
(A7FIL+), the normal reported movement of the hAR
in response to ligand was observed. Unliganded cytoplasmic hAR quickly
translocated to a nuclear site upon addition of ligand (C and D).
Interestingly, the
GFP-hAR559-918 protein
remained in the cytoplasm (E) in the presence or absence of ligand in
M2FIL- cells. In contrast unliganded
GFP-hAR559-918 was
constituitively nuclear in A7FIL+ cells (F), as
previously reported (22). We also observed a shift in morphology in the
A7FIL+ cells, where the cells moved along their
axes to become more spherical in response to ligand. This effect has
been reported previously for the GR (23). In accordance with this
study, there appears to be an underlying nuclear organization of
GFP-hAR accumulation. A known nuclear localizing protein, Tip60 (tat
interacting protein 60) (24) was fused to a GFP to analyze the movement
of a non-steroid hormone receptor construct in the M2 and A7 cell
lines. No aberrant localization of this construct was observed in the
absence of filamin in the absence or presence of mibolerone (G and H).
Taken together, these results suggest that the cytoskeletal protein
filamin is important in the nuclear translocation of the activated
hAR.

View larger version (19K):
[in this window]
[in a new window]
|
Figure 6. hAR Shuttling in a Cell Line Deficient in Filamin
Expression
Confocal micrographs represent GFP-hAR expressing cells. To analyze the
functional significance of the hAR-filamin interaction, hAR nuclear
translocation was studied in a cell line deficient in filamin
expression, denoted (M2FIL-), and the corresponding cell
line with filamin was stably transfected at near wild-type levels
(A7FIL+). In the absence of steroid, the cellular
localization of GFP-hAR1-918 in
M2FIL- and A7FIL+ cell lines is predominantly
cytoplasmic with background nuclear fluorescence (A and C,
respectively). After 20 min exposure to mibolerone, the cellular
localization of the GFP-hAR1-918 construct
remains predominately cytoplasmic in M2FIL- cells (B).
Conversely, in filamin-containing cells (A7fil+) the
cellular location of GFP-hAR1-918 is
predominately nuclear (D). The GFP-hAR559-918
construct, known to be constituitively nuclear even in the absence of
ligand, was predominately cytoplasmic in the M2FIL- cell
line (E). The nuclear translocation of this construct was again
restored in the filamin containing A7FIL+ cell line in the
presence of ligand (F). A control trafficking protein, Tip60, known to
be constituitively nuclear in localization, was unaffected by the
absence of filamin (M2 Fil-) in the absence (G) or
presence (H) of ligand. Scale bar represents 25 µm.
Similar results were observed in two further experiments.
|
|
Filamin Deficiency Affects Ligand-Induced hAR Transcriptional
Activity
The transcriptional activity of GFP-hAR was investigated in
filamin-deficient (M2FIL-) and proficient
(A7FIL+) cell lines (Fig. 7
). Transient transfection of GFP-hAR and
p(ARE)3-Luc into M2FIL-
cells failed to induce a response to the synthetic ligand, Mibolerone.
In contrast, a similar transfection into A7FIL+
cells resulted in a 4.5-fold increase in luciferase activity in
response to ligand. These results demonstrate that GFP-hAR is
functional and that the inability of GFP-hAR to translocate to the
nucleus in filamin-deficient cells results in lower transcriptional
activity.

View larger version (11K):
[in this window]
[in a new window]
|
Figure 7. Filamin-Deficient Cells Exhibit Reduced hAR
Transcriptional Activity
Transient transfection of the GFP-hAR and p(ARE)3-luc
into filamin-deficient M2 cells failed to induce a response to
synthetic ligand, mibolerone. Conversely, similar transfections into
filamin-positive A7 cells exhibited a 4.5-fold increase in luciferase
activity in response to ligand. Graph represents data
from three independent experiments performed in triplicate.
Error bars represent the SD of three
experiments.
|
|
 |
DISCUSSION
|
---|
In this study, we have identified the actin-binding protein
filamin as interacting with the hAR. Filamin, a 280-kDa protein, was
originally identified as a structural component of the cell
cytoskeleton governing the solation-gellation status of the cytoplasm
at the cell membrane (25). This is facilitated by filamins ability to
cross-link F-actin fibers into orthogonal arrays, thereby defining the
cells 3-D structure and motility. Filamin consists of three
distinct domains: 1) an N-terminal actin-binding domain homologous to
those found in other actin-binding proteins such as dystrophin,
vinculin, and talin, 2) a central rod-like domain consisting of 24
repeat regions that forms the ß-sheet backbone of the protein with
domains highly conserved in filamin isoforms, 3) a C-terminal domain
containing the 24th repeat motif that defines a self-association domain
forming the interface of the filamin dimer, creating a leaf spring-like
molecule (26).
Filamin consists of three main isoforms, assigned
, ß, and
. All three share a high degree of sequence homology due to their
role as F-actin cross-linking proteins within the cell. ABP 280 or
-filamin is the original isoform and is located telomeric to the
color vision locus (R/GCP) and centromeric to G6PD in Xq28 (27).
Filamin has previously been identified as interacting with the
ß1-integrins, transmembrane molecules involved in cell-cell contact
and transmembrane signaling (28). The integrins are also critical in
the formation of focal adhesions, local areas on the cell membrane
implicated in signal transduction events from the extracellular matrix
to the cell nucleus. The structure of focal adhesions also links the
integrins with F-actin and actin-associating proteins such as talin,
vinculin, and filamin.
Another interacting partner of filamin is the stress-activated protein
kinase SEK-1, responsible for signal transduction to downstream
molecules JNK and c-Jun affecting cellular responses such as growth and
differentiation (29). It is unclear as to the role of filamin in this
signal transduction pathway, whether filamin itself is phosphorylated
or is acting as an anchoring protein similar in function to JIP-1 (JNK
inhibiting protein), which also allows cross-talk between the
mitogen-activated protein kinase (MAPK) and SAPK pathways (30), remains
to be determined.
More recent studies have identified caveolin-1, a
cholesterol-binding integral membrane protein, as an interacting
partner for filamin (20). The association between androgen-independent
prostate cancer and high expression of caveolin-1 has been documented
(31, 32). The identification of filamin as an hAR interactor highlights
a potential link between caveolin-1, filamin, and the hAR in
androgen-independent prostate cancer.
The melanoma cell line deficient in filamin expression has been well
characterized elsewhere (33, 34). The majority of work on this cell
line analyzed the cell plasma membrane activity relating to filamin
expression. Cells deficient in filamin expression display an increased
occurrence of membrane blebbing due to a decrease in the local actin
polymerization rate. The retraction of these cell surface protrusions
is dependent upon the establishment of a stable actin network, and the
prolonged blebbing displayed by the filamin-deficient cell line is due
to the local rate of actin polymerization being outpaced by the
fluid-driven expansion of the cell membrane. This prolonged blebbing is
abolished when filamin is reexpressed to wild-type levels and increases
the rate of actin polymerization leading to increased protrusive
activity, thus increasing the motility of the cell. The importance of
filamin-dependent motility is highlighted in an X-linked male lethal
condition known as periventricular heterotopia (PH). This condition
arises due to a mutation in the filamin gene resulting in aberrant
neural cell migration within the neural network (35; reviewed in Ref.
36).
This study implicates filamin in the cytoplasmic trafficking of
the hAR. There has been evidence to suggest that SHRs interact with
components of the cytoskeletal architecture, in particular the studies
carried out on the GR (11) that demonstrated an intact cytoskeletal
network is important in the shuttling function of the receptor.
Disruption of the cyto-architecture rendered the GR unable to shuttle
between the cytoplasm and the nucleus. This implies that there is a
regulatory component within the cytoskeleton essential for the
trafficking of molecules to the nucleus. Earlier work (37) presented
the first evidence that SHRs associate with the actin cytoskeleton.
This study demonstrated that the inactive 8S GR contacts the actin
filaments via its binding to Hsp90. Hsp90, similarly to filamin, also
has the ability to cross-link actin filaments (38). Upon hormone
binding, the Hsp90-receptor complex dissociates and the receptor
transforms into the activated complex able to bind
glucocorticoid-responsive elements on target genes within the nucleus.
The conclusions from this study were that the GR (and possibly other
SHRs) bind the actin fibers via their Hsp90 moieties, and this binding
anchors the inactive receptor and prevents translocation to the
nucleus. Later work, again studying GR translocation, implicated the
microtubule network as important in the mechanism of action of GR
hormones (39).
The function of filamin, in the context of hAR cytoplasmic
trafficking, may involve the disruption of the high-affinity
association between Hsp90 and the receptor. In the absence of filamin,
the receptor-Hsp90 complex may remain anchored in an inactive state to
the actin filaments even in the presence of steroid and an available
nuclear localization sequence on the receptor. It is tempting to
speculate that filamin may be acting as a mediator between the receptor
and the molecular chaperone Hsp90 controlling the release of activated
receptor after ligand binding. Interaction between heat shock proteins
and filamin has been detailed (40). The small stress protein cvHsp has
been reported to interact with the C-terminal tail of
-filamin in a
yeast two-hybrid assay. It is plausible that filamin may interact with
Hsp90 in a similar fashion in complex with the hAR.
This study has shown that the filamin-hAR interaction is not steroid
dependent, suggesting receptor anchoring in the absence of ligand. In
the presence of ligand the affinity of the interaction is increased
and, after Hsp90 dissociation, filamin may be acting as a molecular
chaperone, maintaining the active hAR in a stable conformation and
fulfilling its other role as a signaling molecule interacting with
components of the SAPK and MAPK pathways.
This study also has clinical implications. A prostate cancer
susceptibility locus, termed HPCX, has recently been identified in the
region of Xq2728, where the filamin locus maps (41). Linkage analysis
in 153 prostate cancer families over a 30 centimorgan (cM)
region of HPCX has provided additional support for the existence of a
prostate cancer susceptibility locus at Xq28 in a separate study (42).
Our investigations highlight an altered response of the hAR to steroid
in a cell line lacking expression of filamin, which is restored in its
presence. This work provides an insight into hAR movement. Further
investigation is required to ascertain whether this interaction extends
to other members of the SHR superfamily and the significance of this
interaction in clinical specimens.
 |
MATERIALS AND METHODS
|
---|
Yeast Two-Hybrid Analysis
Two-hybrid methodology, bait use, and hAR primer design were
performed as described previously (43). An hAR fragment comprising
residues 559918, encompassing the DNA-binding and ligand-binding
domains, was used as bait in the assay. Positive clones were isolated,
sequenced, and submitted into the GenBank database. One positive clone,
1.002 kb in size, gave a 100% sequence homology to the actin binding
protein filamin (ABP-280, accession no. X53416).
The 1.002-kb filamin clone (hereafter designated
Fil1788-2121) in pACT2 was
transformed back into the yeast strain PJ694A as previously described
(44) with truncated forms of the hAR in pAS21. Transformants were
grown on media lacking leucine and tryptophan, and resultant colonies
were grown in selective media overnight at 30 C in the presence of 1
µM dihydrotestosterone. Samples were diluted to
A600 0.2 and grown to an
A600 of 0.60.8 and split into three 1-ml
samples. Samples were subjected to liquid LacZ assays as described
previously (45). Individual experiments were performed in
triplicate.
In Vitro Interaction of hAR and Filamin
Fil1788-2121cDNA
was excised with EcoRI and HindIII digestion from
pAS21 and directionally cloned into pT77 or pRSETC
(Invitrogen, San Diego, CA) via EcoRI and
HindIII sites. hAR fragments used in the yeast
two-hybrid analysis were excised from pAS21 with EcoRI and
cloned into the EcoRI site of pRSETC. Constructs were
sequenced to confirm the maintenance of the open reading frame.
Template DNA was subjected to Geneclean (Anachem, Luton, UK) to
remove any contaminant RNase, and the coupled Transcription/Translation
Kit (Promega Corp., Madison, WI) was used for in
vitro interaction analysis. Templates were labeled with
35S-methionine and the reaction was performed
according to the manufacturers guidelines. After the 90-min
incubation, samples were split and mixed equally, and 1 ml of reaction
lysis buffer (50 mM Tris, pH 7.5, 150
mM NaCl, 0.2 mM
Na3VO4, 0.5% Nonidet P-40,
1 mM phenylmethlsulfonyl fluoride, 1
mM dithiothreitol, 25 µg/ml leupeptin, 25
µg/ml aprotinin, 25 µg/ml pepstatin) was added. Interacting
proteins were precipitated and visualized as previously described
(43).
Coimmunoprecipitation
Approximately 107 cells from an LNCaP,
hAR-positive cell line were washed in cold PBS, harvested, and lysed in
reaction lysis buffer on ice for 30 min. Lysates were centrifuged at
14,000 x g at 4 C for 10 min. Supernatants were
precleared with 25 µl protein G sepharose (PGS) after three
washes in reaction lysis buffer and rotated at 4 C for 4 h. PGS
and nonspecific bound protein were removed by centrifugation at
14,000 x g for 5 min. Immunoprecipitations were
performed with 4 µg of mouse monoclonal antifilamin antibody
(Chemicon Intl. Inc., Temecula, CA) or 4 µg of rabbit
polyclonal C-terminal AR antibody (Santa Cruz Biotechnology, Inc., Santa Cruz, CA) and incubated overnight at 4 C with
rotation. After incubation, a further 25 µl of PGS were added to
immunoprecipitate samples and returned to 4 C for 1 h with
rotation. PGS with bound protein complexes was recovered by
centrifugation at 14,000 x g for 5 min, and samples
were washed once in wash buffer A (PBS, 0.2% Triton-X-100, 350
mM NaCl) and twice in wash buffer B (PBS, 0.2%
Triton-X-100). Sample buffer containing 10% ß-mercaptoethanol was
added to recovered fractions. Samples were resolved on 10% denaturing
polyacrylamide gels for 30 min and transferred to nitrocellulose
filters. The membrane was probed with polyclonal AR (1:500) or
monoclonal filamin (1:1000) antibodies. Immunoreaction was visualized
using enhanced chemiluminescence (ECL) reagents (Amersham Pharmacia Biotech, Arlington Heights, IL) and developed on
radiographic film.
Deletion Constructs of Filamin Clone
Three deletion constructs of the
Fil1788-2121 in pACT2 were
constructed via PCR. Primers used to generate deletion constructs were:
Fil1788-2081, pACT2FSP
(CLONTECH Laboratories, Inc., Palo Alto, CA), 2R-5'-TGA
GCC CAC CAT AGC CTG C-3';
Fil1930-2081, 1F-5'-GGG
ACT ACA GCA TTC TAG TC-3', 2R-5'-TGA GCC CAC CAT AGC CTG C-3';
Fil1788-1930, pACT2FSP
(CLONTECH Laboratories, Inc.), 1R-5'-GAC TAG AAT GCT GTA
GTC CC-3'. Amplified inserts were cloned into pCR2.1
(Invitrogen) and subcloned into pACT2 via the
EcoRI site. Samples were sequenced to verify the open
reading frame. Deletion constructs were cotransformed with
AR559-918 construct in
pAS21 into yeast strain PJ694A, and resultant transformants were
assayed for ß-galactosidase activity as described previously
(45).
Transient Transfection
COS-7, M2, and A7 cells were grown in steroid-depleted
RPMI 1640 medium containing 10% FCS 48 h before transfection.
Cells were transfected with either pCDNA3-AR, pEGFP-Tip60, pEGFP-hAR,
pCMV-Fil1788-2121, or
pMMP2-Luc (donated by Dr. Y. Sun, Parke-Davis, Ann Arbor,
MI) and p(ARE)3-Luc (provided by Dr. D.
Gioeli, University of Virginia, Charlottesville, VA), as
indicated, using Superfect reagent (QIAGEN, Chatsworth,
CA). A total of 1.5 µg DNA was used per 35- mm well. Transfected
cells were cultured in steroid-depleted medium with or without
synthetic androgen, Mibolerone, where indicated. After 48 h
incubation, cells were harvested, and luciferase activities were
determined using a luciferase reporter system (Promega Corp.). pCMV-ß-gal was used as an internal control for
normalization of transfection efficiency.
Microscopic Analysis of GFP-hAR Chimera in M2 and A7 Cell
Lines
Full-length hAR (corresponding to residues 1918) was cloned
into the XbaI site of the GFP plasmid pEGFP-C1
(CLONTECH Laboratories, Inc.) to generate
GFP-hAR1-918. Residues
559918 of the hAR comprising DNA-binding and ligand-binding domains
of the hAR were cloned into the EcoRI site of pEGFP-C2
(CLONTECH Laboratories, Inc.) to produce
GFP-hAR559-918. Full-
length Tip60 was isolated as described previously (43) and cloned into
the BamHI site of pEGFP-C2 (CLONTECH Laboratories, Inc.). Human epithelial melanoma cell lines, M2 (filamin -ve)
and A7 (filamin +ve) (a kind gift from Dr. T. Stossel,
Harvard University, Cambridge, MA), were used in the GFP-hAR
translocation assays. Cell lines were grown overnight on 22 x 22
mm microscope cover slips in six-well cell culture plates. Cells were
transfected with 2 µg of
GFP-hAR1-918 or
GFP-hAR559-918 construct
using Superfect and cultured in RPMI 1640 medium for 24 h. Cells
were then placed in steroid-depleted media for a further 24 h and
exposed to 10 nM Mibolerone for 0, 5, and 20 min.
Coverslips were washed with sterile PBS and fixed in 100% methanol for
30 min at -20 C, dried, inverted on microscope slides, and mounted in
antifading medium (Shandon Southern Instruments, Inc., Sewickley,
PA). Slides were analyzed on an MRC 600 scanning laser confocal
microscope (Bio-Rad Laboratories, Inc., Richmond, CA).
 |
ACKNOWLEDGMENTS
|
---|
We would like to thank Dr. Trevor Booth for his technical
assistance in obtaining the confocal images, Dr. Tom Stossel for
providing the M2 and A7 cell lines, and Drs. Yi Sun and Dan Gioeli for
the pMMP2-Luc and p(ARE)3-Luc plasmids,
respectively.
 |
FOOTNOTES
|
---|
Address requests for reprints to: Dr. C. N. Robson, Prostate Research Group, School of Surgical and Reproductive Sciences, Medical School, University of Newcastle upon Tyne, Framlington Place, Newcastle upon Tyne, England. NE2 4HH. E-mail: c.n.robson{at}ncl.ac.uk
Received for publication March 31, 2000.
Revision received June 12, 2000.
Accepted for publication July 3, 2000.
 |
REFERENCES
|
---|
-
Evans RM 1988 The steroid and thyroid receptor
superfamily. Science 240:889895[Medline]
-
OMalley BW 1990 The steroid receptor superfamily: more
excitement predicted for the future. Mol Endocrinol 4:363369[Medline]
-
King WJ, Greene GL 1984 Monoclonal antibodies localize
oestrogen receptor in the nuclei of target cells. Nature 307:745747[Medline]
-
Perrot-Applanat M, Logeat M, Groyer-Picard MT, Milgrom E 1985 Immunocytochemical study of mammalian progesterone receptor using
monoclonal antibodies. Endocrinology 116:14731484[Abstract]
-
Wikstrom AC, Bakke O, Okret S, Bronnegard M, Gustafsson JA 1987 Intracellular localization of the glucocorticoid receptor:
evidence for cytoplasmic and nuclear localization. Endocrinology 120:12321242[Abstract]
-
Lombes M, Binart N, Delahaye F, Baulieu EE, Rafestin-Oblin ME 1994 Differential intracellular localization of human
mineralocorticosteroid receptor on binding of agonists and antagonists.
Biochem J 302:191197[Medline]
-
Jenster G, Trapman J, Brinkmann AO 1993 Nuclear import of the
human androgen receptor. Biochem J 293:761768[Medline]
-
Georget JM, Lobaccaro B, Terouanne B, Mangeat P, Nicolas JC,
Sultan C 1997 Trafficking of the androgen receptor in living cells with
fused green fluorescent protein-androgen receptor. Mol Cell Endocrinol 129:1726[CrossRef][Medline]
-
Zhou ZX, Sar M, Simental JA, Lane MV, Wilson EM 1994 A
ligand-dependent bipartite nuclear targeting signal in the human
androgen receptor: requirement for the DNA-binding domain and
modulation by NH2-terminaland carboxyl-terminal sequences. J Biol
Chem 269:1311513123[Abstract/Free Full Text]
-
Perrot-Applanat M, Lescop P, Milgrom E 1992 The cytoskeleton
and the cellular traffic of the progesterone receptor. J Cell Biol 119:337348[Abstract]
-
Galigniana MD, Housley PR, DeFranco DB, Pratt WB 1999 Inhibition of glucocorticoid receptor nucleocytoplasmic shuttling by
okadaic acid requires intact cytoskeleton. J Biol Chem 274:1622216227[Abstract/Free Full Text]
-
Kang KI, Devin J, Cadepond F, Jibard N, Guiochon-Mantel A,
Baulieu EE, Catelli MG 1994 In vivo functional
protein-protein interaction: nuclear targeted hsp90 shifts cytoplasmic
steroid receptor mutants into the nucleus. Proc Natl Acad Sci USA 91:340344[Abstract]
-
Fejes-Toth G, Pearce D, Naray-Fejes-Toth A 1998 Subcellular
localisation of mineralocorticoid receptors in living cells: effects of
receptor agonists and antagonists. Proc Natl Acad Sci USA 95:29732978[Abstract/Free Full Text]
-
Zhu XG, Hanover J, Hager GL, Cheng SY 1998 Hormone-induced
translocation of thyroid hormone receptors in living cells visualized
using a receptor green fluorescent protein chimera. J Biol Chem 273:2705827063[Abstract/Free Full Text]
-
Htun H, Holth L, Walker D, Davie JR, Hager GL 1999 Direct
visualization of the human estrogen receptor alpha reveals a role for
ligand in the nuclear distribution of the receptor. Mol Biol Cell 10:471486[Abstract/Free Full Text]
-
Michigami T, Suga A, Yamazaki M, Shimizu C, Cai G, Okada S,
Ozono K 1999 Identification of amino acid sequence in the hinge region
of human vitamin D receptor that transfers a cytosolic protein to the
nucleus. J Biol Chem 274:3353133538[Abstract/Free Full Text]
-
Lim CS, Baumann CT, Htun H, Xian W, Irie M, Smith CL, Hager GL 1999 Differential localisation and activity of the A-and B-forms of the
human progesterone receptor using green fluorescent protein chimeras.
Mol Endocrinol 13:366375[Abstract/Free Full Text]
-
Fields S, Song O 1989 A novel genetic system to detect
protein-protein interactions. Nature 340:245246[CrossRef][Medline]
-
Xu WF, Xie Z, Chung DW, Davie EW 1998 A novel human
actin-binding protein homologue that binds to platelet glycoprotein
Ib
. Blood 92:12681276[Abstract/Free Full Text]
-
Stahlhut M, van Deurs B 2000 Identification of filamin as a
novel ligand for caveolin-1: evidence for the organization of
caveolin-1-associated membrane domains by the actin cytoskeleton. Mol
Biol Cell 11:325337[Abstract/Free Full Text]
-
Leonardi A, Ellinger-Ziegelbauer H, Franzoso G, Brown K,
Siebenlist U 2000 Physical, functional interaction of filamin
(actin-binding protein-280), tumor necrosis factor receptor-associated
factor 2. J Biol Chem 275:271278[Abstract/Free Full Text]
-
Simental JA, Sar M, Lane MV, French FS, Wilson EM 1991 Transcriptional activation and nuclear targeting signals of the human
androgen receptor. J Biol Chem 266:510518[Abstract/Free Full Text]
-
Htun H, Barsony J, Renyi I, Gould DL, Hager GH 1996 Visualisation of the glucocorticoid receptor translocation and
intranuclear organisation in living cells with a green fluorescent
protein chimera. Proc Natl Acad Sci USA 93:48454850[Abstract/Free Full Text]
-
Yamamoto T, Horikoshi M 1997 Novel substrate specificity of
the histone acetyltransferase activity of HIV-1 tat interactive protein
Tip60. J Biol Chem 272:3059530598[Abstract/Free Full Text]
-
Koteliansky VE, Shirinsky V, Gneushev GN, Smirnov 1981 Filamin, a high relative molecular mass actin-binding protein from
smooth muscles, promotes actin polymerization. FEBS Lett 136:98100[CrossRef][Medline]
-
Gorlin JB, Yamin R, Egan S, Stewart M, Stossel TP, Kwiatkowski
DJ, Hartwig JH 1990 Human endothelial actin-binding protein (ABP-280,
nonmuscle filamin): a molecular leaf spring. J Cell Biol 111:10891105[Abstract]
-
Gorlin JB, Henske E, Warren ST, Kunst CB, DUrso M, Palmiera
G, Hartwig JH 1993 Actin-binding protein (ABP-280) filamin gene (FLN)
maps telomeric to the color vision locus (R/GCP) and centromeric to
G6PD in Xq28. Genomics 17:496498[CrossRef][Medline]
-
Loo DT, Kanner S, Aruffo A 1998 Filamin binds to the
cytoplasmic domain of the beta1-integrin Identification of amino acids
responsible for this interaction. J Biol Chem 273:2330423312[Abstract/Free Full Text]
-
Marti A, Luo Z, Cunningham C, Ohta Y, Hartwig J, Stossel TP,
Kyriakis JM, Avruch J 1997 Actin-binding protein-280 binds the
stress-activated protein kinase (SAPK) activator SEK-1 and is required
for tumor necrosis factor-
activation of SAPK in melanoma cells.
J Biol Chem 272:26202628[Abstract/Free Full Text]
-
Pedram A, Razandi M, Levin ER 1998 Extracellular
signal-regulated protein kinase/jun kinase cross-talk underlies
vascular endothelial cell growth factor induced endothelial cell
proliferation. J Biol Chem 273:2672226728[Abstract/Free Full Text]
-
Yang G, Truong L, Wheeler TM, Thompson TC 1999 Caveolin-1
expression in clinically confined human prostate cancer: a novel
prognostic marker. Cancer Res 59:57195723[Abstract/Free Full Text]
-
Nasu Y, Timme T, Yang G, Bangma CH, Li L, Ren C, Park SH,
DeLeon M, Wang J, Thompson TC 1998 Suppression of caveolin expression
induces androgen sensitivity in metastatic androgen-insensitive mouse
prostate cancer cells. Nat Med 4:10621064[CrossRef][Medline]
-
Cunningham CC, Gorlin JB, Kwiatkowski DJ, Hartwig JH, Janmey
PA, Byers HR, Stossel TP 1992 Actin-binding protein requirement for
cortical stability and efficient locomotion. Science 255:325327[Medline]
-
Cunningham CC 1995 Actin polymerization and intracellular
solvent flow in cell surface blebbing. J Cell Biol 129:15891599[Abstract]
-
Fox JW, Lamperti E, Eksioglu YZ, Hong SE, Feng Y, Graham DA,
Scheffer IE, Dobyns WB, Hirsch BA, Radtke RA, Berkovic SF, Huttenlocher
PR, Walsh CA 1998 Mutations in filamin 1 prevent migration of cerebral
cortical neurons in human periventricular heterotopia. Neuron 21:13151325[Medline]
-
Fox JW, Walsh C 1999 Periventricular heterotopia and the
genetics of neuronal migration in the cerebral cortex. Am J Hum
Genet 65:1924[CrossRef][Medline]
-
Miyata Y, Yahara I 1991 Cytoplasmic 8S glucocorticoid receptor
binds to actin filaments through the 90-kDa heat shock protein moiety.
J Biol Chem 266:87798783[Abstract/Free Full Text]
-
Koyasu S, Nishida E, Kadowaki T, Matsuzaki F, Iida K, Harada
F, Kasuga M, Sakai H, Yahara I 1986 Two mammalian heat shock proteins,
HSP90 and HSP100, are actin-binding proteins. Proc Natl Acad Sci USA 83:80548058[Abstract]
-
Akner G, Wikstrom A, Gustafsson JA 1995 Subcellular
distribution of the glucocorticoid receptor and evidence for its
association with microtubules. J Steroid Biochem Mol Biol 52:116[CrossRef][Medline]
-
Krief S, Faivre J, Robert P, Le Douarin B, Brument-Larignon N,
Lefrere I, Bouzyk MM, Greller LD, Tobin FL, Souchet M, Bril A 1999 Identification and characterization of cvHsp A novel human small stress
protein selectively expressed in cardiovascular and insulin-sensitive
tissues. J Biol Chem 274:3659236600[Abstract/Free Full Text]
-
Xu J, Meyers D, Freije D, Isaacs S, Wiley K, Nusskern D, Ewing
C, Wilkens E, Bujnovszky P, Bova GS, Walsh P, Isaacs W, Schleutker J,
Matikainen M, Tammela T, Visakorpi T, Kallioniemi OP, Berry R, Schaid
D, French A, McDonnell S, Schroeder J, Blute M, Thibodeau S, Trent J,
et al. 1998 Evidence for a prostate cancer susceptibility
locus on the X chromosome. Nat Genet 20:175179[CrossRef][Medline]
-
Lange EM, Chen H, Brierley K, Perrone EE, Bock CH, Gillanders
E, Ray ME, Cooney KA 1999 Linkage analysis of 153 prostate cancer
families over a 30-cM region containing the putative susceptibility
locus HPCX. Clin Cancer Res 5:40134020[Abstract/Free Full Text]
-
Brady ME, Ozanne DM, Gaughan L, Waite I, Cook S, Neal DE,
Robson, CN 1999 Tip60 is a nuclear hormone receptor co-activator.
J Biol Chem 274:1759917604[Abstract/Free Full Text]
-
Gietz RD, St-Jean JA, Woods RA, Schiestl RH 1992 Improved
method for high efficiency transformation of intact yeast cells.
Nucleic Acids Res 20:1425[Medline]
-
Gietz RD, Triggs-Raine B, Robbins A, Graham KC, Woods RA 1997 Identification of proteins that interact with a protein of interest:
applications of the yeast two-hybrid system. Mol Cell Biochem 172:6779[CrossRef][Medline]