Download 1 T-cadherin is located in the nucleus and centrosomes in

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Cytosol wikipedia , lookup

Tissue engineering wikipedia , lookup

Signal transduction wikipedia , lookup

Extracellular matrix wikipedia , lookup

Endomembrane system wikipedia , lookup

Cell growth wikipedia , lookup

SULF1 wikipedia , lookup

Cell encapsulation wikipedia , lookup

Cell cycle wikipedia , lookup

Cell culture wikipedia , lookup

Cellular differentiation wikipedia , lookup

Cytokinesis wikipedia , lookup

Cell nucleus wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

Mitosis wikipedia , lookup

List of types of proteins wikipedia , lookup

Amitosis wikipedia , lookup

Transcript
Articles in PresS. Am J Physiol Cell Physiol (September 2, 2009). doi:10.1152/ajpcell.00237.2009
1
T-cadherin is located in the nucleus and centrosomes in endothelial cells.
2
3
Alexandra V. Andreeva1*, Mikhail A. Kutuzov1, Vsevolod A. Tkachuk2, Tatyana A. Voyno-
4
Yasenetskaya1*
5
6
1
Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA.
7
2
Cardiology Research Center, Moscow, Russia.
8
9
* Corresponding authors. Deparment of Pharmacology (MC 868), University of Illinois at
10
Chicago, 909 S. Wolcott Ave. COMRB, Chicago, IL 60612, USA.
11
Tel.: +1 312 996 9823; fax: +1 312 996 1225.
12
E-mail addresses: [email protected] (A. V. Andreeva), [email protected] (T. A. Voyno-
13
Yasenetskaya).
14
15
Running title: Nuclear and centrosomal location of T-cadherin
16
1
Copyright © 2009 by the American Physiological Society.
17
ABSTRACT
18
19
T-cadherin (H-cadherin, cadherin 13) is upregulated in vascular proliferative disorders and in
20
tumor-associated neovascularization, and is deregulated in many cancers. Unlike canonical
21
cadherins, it lacks transmembrane and intracellular domains and is attached to the plasma
22
membrane via a glycosylphosphatidylinositol anchor. T-cadherin is thought to function in
23
signaling rather than as an adhesion molecule. Some interactive partners of T-cadherin at the
24
plasma membrane have recently been identified. We examined T-cadherin location in human
25
endothelial cells using confocal microscopy and subcellular fractionation. We found that a
26
considerable proportion of T-cadherin is located in the nucleus and in the centrosomes. T-
27
cadherin colocalized with a centrosomal marker γ-tubulin uniformly throughout the cell cycle at
28
least in human umbilical vein endothelial cells. In the telophase, T-cadherin transiently
29
concentrated in the midbody and was apparently degraded. Its overexpression resulted in an
30
increase in the number of multinuclear cells, while its downregulation by siRNA led to an
31
increase in the number of cells with multiple centrosomes. These findings indicate that
32
deregulation of T-cadherin in endothelial cells may lead to disturbances in cytokinesis or
33
centrosomal replication.
34
35
Key words: cadherin family, cell cycle, cytokinesis, GPI anchor, HPAEC.
36
37
2
38
INTRODUCTION
39
T-cadherin is a glycoprotein that belongs to the cadherin cell adhesion family. In contrast to
40
canonical cadherins, T-cadherin lacks many amino acids crucial for Ca2+-dependent intercellular
41
dimerization (15, 60). As a consequence, it is monomeric in the absence and in the presence of
42
calcium (15) and is thought to mainly function as a signaling molecule. Another unique feature
43
of T-cadherin is the absence of the transmembrane and cytoplasmic domains (although a rare
44
cell line-specific transmembrane form has been reported (54)). T-cadherin is anchored within
45
lipid rafts through glycosylphosphatidylinositol (GPI) (60, 68) and may possibly use cytoplasmic
46
domains of several interactive partners (58) to transduce signals inside the cell. T-cadherin may
47
operate through integrin-linked kinase (ILK), which is upstream of the Akt / GSK3β / β-catenin
48
pathway (41, 42). It has been suggested and confirmed in an in vivo model that T-cadherin may
49
play an antiapoptotic and protective role under hypoxic conditions (30, 42).
50
In confluent endothelial cells (ECs) T-cadherin, unlike VE-cadherin (which is mainly present at
51
the cell-cell junctions), is located over the entire cell body with only a slight enrichment at cell
52
borders, while in wounded cultures it is polarized to the leading edge of migrating cells (59).
53
Some recent reports described nuclear localization of cleaved cytoplasmic domains of canonical
54
cadherins (21, 66) and their potential role in the modulation of gene transcription (21).
55
Several lines of evidence point to a potential involvement of T-cadherin in cell cycle regulation in
56
different cell lines. T-cadherin expression is decreased or undetectable in some tumor samples
57
and various cancer cell lines (63, 73). In contrast, overexpression of T-cadherin was
58
demonstrated in tumorigenic liver tissue and a hepatocellular carcinoma cell line (63). The role
59
of T-cadherin in tumor neovascularization was recently confirmed using in vivo model (30). T-
60
cadherin expression in T-cadherin-deficient C6 glioma cells results in G2 phase arrest and
61
aneuploidy, which is dependent on increased expression of p21CIP1 and is eliminated in p21CIP1-
62
deficient fibroblasts (35). Ectopic expression of Cdh1, one of the substrate recognition
3
63
components (E3 ligase) of the anaphase promoting complex, stimulates T-cadherin degradation
64
(5). In human umbilical vein endothelial cells (HUVECs), overexpression of T-cadherin leads to
65
an increased expression of cyclin D1, a key regulator of G1 to S-phase progression (24), and to
66
a rapid entrance into S-phase (38).
67
Here we report that in ECs a considerable proportion of T-cadherin is located in the nucleus. It
68
is also present in the centrosomes and co-localizes with γ-tubulin. In the telophase, T-cadherin
69
transiently concentrates in the midbody. Overexpression of T-cadherin results in an increase in
70
the number of multinuclear cells, while its downregulation leads to an increased number of cells
71
with multiple centrosomes. These findings indicate that T-cadherin is a nuclear and centrosomal
72
protein and that its deregulation may interfere with the cell cycle via disturbance in cytokinesis
73
or centrosomal replication.
74
75
MATERIALS AND METHODS
76
Materials. The following antibodies were used: Akt1, Bcl2, calreticulin, GFP, GRP78, LAMP1, T-
77
cadherin (sc-7940; designated here as SC), γ-tubulin, VE-cadherin, HRP-conjugated anti-goat
78
antibody (Santa Cruz Biotechnology); FLAG (M2; Sigma); histone 1 (AE-4; Millipore), GM130,
79
Hsp90, PP5, Rab5, Rac1 (BD Transduction Laboratories), ZO-1 (Zymed). HRP-conjugated anti-
80
mouse and anti-rabbit secondary antibodies were from Amersham. Phalloidin, AlexaFluor 488,
81
594, or 633 anti-mouse, anti-rabbit, or anti-goat antibodies were from Molecular Probes. The
82
affinity-purified polyclonal T-cadherin antibody designated here as TK was described previously
83
(70). FLAG-tagged T-cadherin constructs were kindly provided by Drs Kalpana Ghoshal and
84
Samson Jacob (Ohio State University) (5). T-cadherin constructs and specificity of the
85
antibodies used in this work are depicted in Fig. 1A and Supplementary Fig. 1A.
4
86
Cell culture. HEK 293A cells were cultured in Dulbecco's modified Eagle's medium (DMEM),
87
supplemented with 10% fetal bovine serum. Human endothelial cells were obtained at passage
88
3 from Lonza and cultured as described previously (4). Transient transfection of HEK 293A and
89
ECs was performed using Lipofectamine 2000 (Invitrogen) and SuperFect (Qiagen),
90
respectively, according to manufacturers’ instructions.
91
Confocal microscopy. Cells cultured on gelatin or fibronectin-coated coverslips were fixed with
92
3.7% paraformaldehyde or with 50% methanol (for preservation of centrosomes), followed by
93
permeabilization in 0.5% Triton X-100. Cells were incubated with primary antibodies followed by
94
incubation with appropriate secondary antibodies, using Tris buffered saline containing bovine
95
serum albumin as a blocking buffer.
96
siRNA-mediated depletion. Inhibition of T-cadherin expression was performed using siRNA
97
designed by Dharmacon. Cells were transfected using siRNA transfection reagent (Santa Cruz
98
Biotechnology) or Lipofectamine 2000 (Invitrogen). Control siRNA was purchased from Santa
99
Cruz Biotechnology.
100
Nuclear fractionation. Cells from a 10 cm dish were washed twice with PBS, recovered by
101
scraping and resuspended in 300 μl of lysis buffer, containing 20 mM Tris-HCl, pH 7.4, 100 mM
102
NaCl, 10 mM EGTA, 5 mM MgCl2, 0.5% Na deoxycholate, 0.5% Nonidet P-40, protease
103
inhibitor cocktail (Sigma, 1:200 dilution). Cells were left on ice for 15 min and centrifuged at
104
1000 g for 5 min. Completeness of cell lysis was verified microscopically. The supernatant
105
(“non-nuclear” fraction) was removed and the pelleted nuclei were washed twice in lysis buffer.
106
Isolation of centrosomes. Centrosomes isolation was performed essentially according to ref.
107
(77). Briefly, confluent HUVECs on four 10 cm plates were treated with cytochalasin D (1 μg/ml)
108
and 0.2 μM nocodazol for 1 h at 370C. Cells were collected in 8 ml hypotonic lysis buffer (1mM
109
HEPES, pH 7.2, 0.5% NP-40, 0.5 mM MgCl2, 1 mM DTT, protease and phosphatase inhibitors
5
110
(Sigma, 1:200 dilution)). Nuclei were pelleted by centrifugation at 2,500 g for 10 min. The
111
supernatant was underlaid with a 60% sucrose cushion (0.8 ml), and centrosomes were
112
sedimented at 25,000 g for 30 min. The cushion and two volumes of the buffer above it (2.4 ml
113
total) were collected, mixed and laid on the top of a discontinuous sucrose gradient (1 ml 70%,
114
0.6 ml 50%, 0.6 ml 40% sucrose) and centrifuged at 100,000 g for 1 h. Fractions of 0.45 ml
115
were collected and diluted to reduce sucrose concentration to <20%. Centrosomes were
116
pelleted at 25,000 g for 30 min and dissolved in the SDS sample buffer for electrophoresis.
117
Immunoprecipitaiton and Western blotting. Cells were lysed in 50 mM HEPES, pH 7.5, 50 mM
118
NaCl, 5 mM MgCl2, and 1% Triton X-100. Proteins were immunoprecipitated with appropriate
119
antibodies (as specified in figure legends) and protein A/G agarose (Santa Cruz Biotechnology)
120
for 4 h at 4°C. Immunoprecipitates were washed 3 times with the lysis buffer. Proteins were
121
separated on 5–20% gradient SDS gels and transferred onto a PVDF (Osmonics) or
122
nitrocellulose (Protran, Shchleicher & Schull) membranes. Membranes were probed with
123
appropriate antibodies and developed using Dura reagents (Pierce). Densitometry of protein
124
bands was performed on scanned images using NIH Image 1.63 software.
125
126
127
RESULTS
128
T-cadherin is located in the nucleus in ECs. In accordance with a report that T-cadherin is only
129
slightly enriched at the cell borders (59), we only occasionally observed cell surface staining in
130
the primary cultures of ECs (Fig. 1B, indicated with arrowheads in the upper panel). On the
131
contrary, a robust staining of the nuclei was observed using two different polyclonal T-cadherin
132
antibodies, designated here as TK (raised against synthetic peptides of the first extracellular
133
subdomain (70)) and SC (raised against a peptide overlapping with CD2 and CD3 domains, Fig.
6
134
1A). Nuclear staining was observed in three different cell lines (Fig 1B): human pulmonary
135
artery endothelial cells (HPAECs), human umbilical vein endothelial cells (HUVECs) and human
136
microvascular endothelial cells (HMVECs). It was not due to the secondary antibody used in
137
these experiments, since omitting primary antibodies resulted in the absence of nuclear staining
138
(Fig. 1B, bottom panel).
139
To confirm the nuclear location of T-cadherin, we fractionated ECs into a nuclear fraction and a
140
fraction containing other cellular compartments and cytoplasmic proteins. The fractionation was
141
confirmed using histone H1 as a nuclear marker (Fig. 2 A, B). Approximately half of total
142
endogenous T-cadherin was found in the nuclear fractions in HPAECs, as revealed by Western
143
blotting with the TK antibody, which recognizes 105 and 130 kDa forms (Fig. 2A). A lower
144
proportion (10-20%) of endogenous T-cadherin was found in the nuclear fraction of HUVECs
145
(Fig. 2B). The absence of contamination of the nuclear fractions by proteins present in the post-
146
nuclear supernatant was evident from reprobing the membranes for a number of markers for
147
different subcellular compartments: Bcl2 (mitochondria), calreticulin (ER), GRP78 (ER/PM),
148
LAMP1 (lysosomes), VE-cadherin (adherens junctions), GM130 (Golgi), Hsp90 (cytoplasm),
149
Rab5 (early endosomes), ZO-1 (tight junctions). It is worth noting that some of the examined
150
proteins have been reported to either be present in the nucleus (PP5 (9), ZO-1 (27), Akt1 (56),
151
Rac1 (50, 51)), or to be functional or interacting partners of T-cadherin (Akt1 (42), Rac1 (57),
152
GRP78 (58)). Only trace amounts (except ZO-1 in HPAEC), if any, of these proteins could be
153
detected in the nuclear fractions (Fig. 2C). These data effectively rule out a possibility that the
154
presence of T-cadherin in the nuclear fractions of HPAECs and HUVECs is due to non-specific
155
contamination by proteins from post-nuclear supernatant.
156
The predicted molecular mass of the mature protein without signal peptide is approximately 65
157
kDa (60). In ECs, there are two major forms of T-cadherin, mature 105 kDa and a partially
158
processed precursor 130 kDa, both of them are presumably glycosylated (70). The results of
7
159
subcellular fractionation clearly indicate that both forms of T-cadherin are present in the nucleus.
160
Both T-cadherin antibodies gave similar nuclear patterns in immunofluorescence experiments
161
(Fig. 1B). The SC antibody has not been used in fractionation experiments (Fig. 2), since on
162
Western blots it failed to recognize full length T-cadherin, although it recognized truncated T-
163
cadherin mutant ΔC3-4 with high efficiency (Supplementary Fig. 1A-C). Since T-cadherin has
164
several potential glycosylation sites, this might be due to conformation sensitivity or epitope
165
masking by glycosyl group(s). Since the presence of T-cadherin in the nucleus was highly
166
unexpected and of a considerable potential interest, we also confirmed this finding using
167
polyclonal antibodies against CD1 and CD5 domains of T-cadherin (40), respectively
168
(Supplementary Fig. 1D).
169
Analysis of T-cadherin primary structure did not reveal any canonical nuclear localization
170
sequences, although it is enriched in basic residues and is predicted to be a nuclear protein by
171
the ESLPred (8) and SubLoc (28) algorithms. We also found that T-cadherin contains a Leu-rich
172
sequence LRFSLPSVLLLSLFSLACL within its C-terminal recognition sequence (CSR) that
173
perfectly matches a nuclear export signal consensus Lx2-3Lx2-3LxL (46). Before mature GPI-
174
anchored proteins are delivered to the cell surface, they undergo processing in the ER. Their
175
CSRs are removed by proteolytic cleavage in the ER simultaneously with the attachment of the
176
GPI-anchor to the newly exposed C-terminus (22). We examined the localization of FLAG-
177
tagged T-cadherin construct, where the epitope is placed after the CSR for GPI-attachment (see
178
Supplementary Fig. 1A). C-terminal epitope tags attached after CSRs are not expected to
179
impede the protein proper processing, and are removed when the protein is processed in the
180
ER (2, 7, 65), thus only the molecules that are not (yet) processed are detectable with the
181
epitope-specific antibody. The non-processed FLAG-tagged T-cadherin could be exclusively
182
detected in the non-nuclear fractions (Fig. 2B, Supplementary Fig. 1C), presumably in the ER,
8
183
taken into account a characteristic reticular pattern observed with FLAG-tagged T-cadherin
184
(Supplementary Fig. 1C).
185
We also examined the effect of leptomycin B (LMB, an inhibitor of CRM1-dependent nuclear
186
export) on distribution of endogenous T-cadherin in HUVECs. The proportion of both
187
endogenous T-cadherin forms increased by 1.5-2 fold in the nucleus upon LMB treatment,
188
indicating that the presence of T-cadherin in the nucleus is dynamically regulated. As it is
189
unlikely that mature T-cadherin possesses this CSR sequence, it is possible that it interacts with
190
a partner with functional nuclear export signal.
191
T-cadherin is present in the centrosomes. In dividing HPAECs, T-cadherin was associated with
192
two punctate structures located on the opposite sides of chromatin and thus resembling
193
centrosomes (see Fig. 1B, lower HPAEC panel). Z-sectioning of individual cells confirmed that
194
each mitotic cell contained exactly two T-cadherin-positive structures (Supplementary Fig. 2A),
195
consistent with their identification as centrosomes. Therefore, we examined whether T-cadherin
196
would co-localize with a centrosomal marker γ-tubulin. We found a clear co-localization between
197
the two proteins throughout the cell cycle (Fig. 3; methanol fixation was used in this experiment
198
to optimize centrosome preservation, which is not optimal for visualization of the nuclei). This
199
indicated that T-cadherin is indeed present in the centrosomes. In the telophase it was often
200
transiently concentrated in the midbody, with reduced or undetectable presence in the
201
centrosomes (note that the brightness of the green channel is increased in the telophase panel
202
as compared to other panels in Fig. 3). In HUVECs, these T-cadherin-positive centrosomes
203
could be observed throughout the cell cycle, including mitosis. In HPAECs, a qualitatively similar
204
pattern was observed, however centrosomal T-cadherin staining was most obvious in
205
metaphase and anaphase.
206
Centrosome-associated
207
(Supplementary Fig. 2B), although less efficiently than with the SC antibody (while both
T-cadherin
could
also
9
be
detected
using
the
TK
antibody
208
antibodies stain the nuclei similarly, Fig. 1B). Lower efficiency of the TK antibody might be due
209
to partial unavailability of its epitope (CD1). In PC12 cells, T-cadherin was reported to be
210
ubiquitinated by Cdh1, a component of a major ubiquitination system that controls the
211
proteasome-dependent destruction of cell cycle regulators (5). Proteasomes are located both in
212
the nucleus and in the centrosomes (see Refs in (16)). Cdh1 localizes to the nucleus during
213
interphase, and to the centrosomes during metaphase and anaphase (78). This prompted us to
214
test whether the levels of T-cadherin in HUVECs would be affected by proteasome inhibition.
215
After 4 h incubation with proteasome inhibitors I1 and MG132, the levels of T-cadherin were
216
significantly increased, while incubation with I2 (calpain inhibitor that does not affect proteasome)
217
resulted in only marginal if any increase in T-cadherin levels (Fig. 4A). These data indicate that
218
T-cadherin levels in HUVECs are under control of proteasome-dependent degradation.
219
To confirm the centrosomal location of T-cadherin, we isolated centrosomal fractions from
220
HUVECs by differential centrifugation using a 20-70% discontinuous sucrose gradient. The
221
fractionation was confirmed using γ-tubulin as a centrosomal marker. Immunoblotting showed a
222
biphasic distribution of γ-tubulin (Fig. 4B). The lower molecular weight form, but not the 130 kDa
223
form of T-cadherin could be detected in the centrosomal fractions using the TK antibody and
224
was mainly associated with the first of the two γ-tubulin peaks (Fig. 4B). Reprobing the same
225
blot with the SC antibody (which does not recognize full length glycosylated T-cadherin on
226
Western blots, but may efficiently recognize truncated form(s), see above) revealed the
227
presence of a ~60 kDa band, associated with the second γ-tubulin peak and also found in a
228
denser fraction (Fig. 4B). Thus, the results of cenrosome isolation indicate that only the mature
229
processed form but not the 130 kDa form of T-cadherin is associated with a subfraction of the
230
centrosomes. Our data are also compatible with a truncated form or non-glycosylated T-
231
cadherin present in a distinct centrosomal subfraction.
10
232
To examine whether T-cadherin and γ-tubulin might physically associate, we performed
233
immunoprecipitation from HUVECs. We first attempted to assess whether endogenous T-
234
cadherin might associate with γ-tubulin. However, endogenous T-cadherin could not be
235
detected in γ-tubulin immunoprecipitates, while reciprocal immunoprecipitation proved
236
inconclusive due to nearly identical molecular weight of γ-tubulin and the heavy immunoglobulin
237
chain (data not shown). Taking into account that only a small proportion of endogenous T-
238
cadherin is located in the centrosomes in comparison with the nucleus in ECs, we next
239
attempted to use overexpressed T-cadherin to assess the possibility of its interaction with γ-
240
tubulin. Since the SC antibody recognizes the ΔC3-4 T-cadherin construct, but not full length
241
constructs on Western blots (as discussed above and shown in Supplementary Fig. 1B),
242
HUVECs were transfected with the latter construct or empty vector. Immunoprecipitation was
243
performed using γ-tubulin- and GFP-specific antibodies. The presence of a band of the
244
expected molecular size (see Supplementary Fig. 1B) recognized by the SC antibody could be
245
detected in the material precipitated from T-cadherin transfected cells with the γ-tubulin antibody,
246
but not in the negative controls (Fig. 4C). No corresponding signal could be detected in the
247
immunoprecipitates using FLAG antibody, consistent with a form that has been C-terminally
248
processed in the ER.
249
To assess whether the centrosomal and nuclear location of T-cadherin is specific for ECs, we
250
examined localization of endogenous T-cadherin in HEK 293A cells. Immunoblotting analysis
251
showed that HEK 293A (as well as COS-7) cells express considerably lower levels of T-
252
cadherin relative to total protein than ECs (data not shown). We found that in HEK 293A cells,
253
most of endogeneous T-cadherin co-localized with γ-tubulin and thus was associated with the
254
centrosomes (which are unusually large structures in this cell line (55, 76)), with only very faint
255
nuclear and cell surface staining (Supplementary Fig. 3A). In an attempt to detect association of
256
endogenous T-cadherin with γ-tubulin, we performed immunoprecipitation experiments from
11
257
HEK 293A cells. A ~60 kDa band could be detected in γ-tubulin immunoprecipitates, but not in
258
GFP antibody immunoprecipitates used as a negative control (Supplementary Fig 3B). The
259
molecular weight of this band is close to that expected for non-glycosylated T-cadherin without
260
prepeptide (65 kDa).
261
Interference with T-cadherin expression leads to defects in cell division. As described above, T-
262
cadherin was often transiently concentrated in the midbody, which plays a key role in
263
cytokinesis (6). This observation suggested that T-cadherin might be involved in cytokinesis.
264
Cytokinesis failure leads to appearance of cells containing more than one nucleus. To assess
265
whether overexpression or downregulation of T-cadherin might affect the probability of failed
266
cytokinesis, we examined the proportion of bi- or multinuclear cells among HUVECs transfected
267
with either T-cadherin cDNA or T-cadherin siRNA. A small proportion of ECs is naturally bi- or
268
multinuclear, and an increase in the proportion of such cells has been associated with some
269
physiological or pathological conditions (19, 36, 69). In line with these reports, we observed that
270
primary HUVEC cultures contained 4-5 % cells with more than one nucleus (of which absolute
271
majority were binuclear cells).
272
T-cadherin could be efficiently overexpressed in ECs (Fig. 5A; Supplementary Fig. 1C),
273
resulting in an overall increase in T-cadherin content of up to 10-20 fold (ECL quantification data
274
not shown). Immunofluorescence microscopy revealed that in some cells T-cadherin was
275
present in a reticular pattern (Fig. 5B, upper panel), probably reflecting its predominant
276
presence in the endoplasmic reticulum, while in other cells punctate cytoplasmic as well as
277
nuclear staining was observed (Fig. 5B, middle panel). The proportion of cells containing more
278
than one nucleus was increased among the cells overexpressing T-cadherin (Fig. 5 B, C).
279
Similar results were obtained using overexpression of FLAG-tagged T-cadherin in HUVECs
280
(data not shown) and in HEK 293A cells (Supplementary Fig. 3 C-E).
12
281
We also depleted endogenous T-cadherin in ECs using siRNA approach, and examined the
282
cells for any apparent defects associated with cell division. Both 105 and 130 kDa forms of T-
283
cadherin could be efficiently downregulated in HPAECs and in HUVECs (Fig. 6A). It was
284
reported that the levels of different cadherins may influence each other, for example, VE-
285
cadherin expression is reduced in N-cadherin-deficient endothelium (48)). VE-cadherin levels
286
remained unchanged in the cells with 80-90% T-cadherin depleted (Fig. 6A). In some
287
experiments, 95-98% T-cadherin depletion was achieved, which was accompanied by a 30-40%
288
decrease in VE-cadherin levels (data not shown). T-cadherin depletion also resulted in a
289
considerable decrease in T-cadherin positive staining in immunofluorescence experiments (Fig
290
6B, C).
291
T-cadherin depletion led to an only marginal increase in the proportion of multinucleated cells,
292
which was not statistically significant (Fig. 6E). However, we noticed that downregulation of T-
293
cadherin was accompanied by an increased proportion of mononuclear ECs with multiple
294
centrosomes (Fig. 6 C,D,F), which suggests a specific role of T-cadherin in the control of
295
centrosomal organization.
296
297
DISCUSSION
298
We report here that a considerable proportion of T-cadherin is located in the nucleus, as
299
evidenced by immunofluorescence and by immunoblotting upon cell fractionation using
300
antibodies raised against different epitopes. Nuclear localization of cleaved intracellular domains
301
of some other members of the cadherin family, such as protocadherins α (18), γ (29), Fat1 (33,
302
49), E-cadherin (21) was reported. However, T-cadherin has no intracellular domain and is
303
present in the nucleus as a full-length molecule; therefore trafficking pathways suggested for
304
other cadherins may not be relevant in the case of T-cadherin. Full length cell surface receptors,
305
both G-protein coupled receptors and tyrosine kinase receptors, were reported to translocate to
13
306
the nucleus (17, 20, 25, 53, 62, 72), including LPA1, for which caveolin-mediated endocytosis
307
was suggested (26). Nucleus is rich in lipid environments, which contain lipid raft components,
308
including caveolin-1 (1). However, several lines of evidence argue against a role of caveolin in
309
the nuclear trafficking of T-cadherin. (i) Pertussis toxin inhibits LPA1 trafficking to the nucleus
310
(26), but does not affect the nuclear localization of T-cadherin (our unpublished observations),
311
which makes a common trafficking mechanism unlikely. (ii) Although caveolae-dependent
312
endocytosis was originally considered for GPI proteins (3), later studies suggested a caveolae-
313
independent pinocytotic pathway (44, 45, 64) or macropinocytotic pathways triggered by a
314
clustering agent or being ligand-independent (14, 74). (iii) In ECs, caveolin is not involved in T-
315
cadherin-mediated signaling (59). However, a recently described T-cadherin interaction with
316
GRP78 (58), which is reportedly involved in caveolae-dependent internalization and nuclear
317
localization of extracellular matrix protein DMP1 (61), reintroduces the question about a possible
318
role of caveolae.
319
In some cell lines, T-cadherin may be not GPI-modified, but rather expressed as a protein with a
320
transmembrane domain (54). Thus, it cannot be excluded that precise sites of processing of T-
321
cadherin in ECs might differ from those assumed on the basis of canonical processing of GPI-
322
anchored proteins.
323
If T-cadherin is processed as a canonical GPI-anchored protein, the questions arise whether it
324
is targeted to the nucleus after its delivery to the PM, or directly from the ER. Some models for
325
the nuclear translocation of cell surface receptors have been suggested (47), and although
326
these questions are beyond the scope of this work, T-cadherin would be an excellent model to
327
study this process.
328
Apart from the question of how T-cadherin is imported into the nucleus, another question is what
329
functional role the nuclear T-cadherin may play. Cleaved intracellular domain of E-cadherin may
330
modulate gene transcription (21). Some other transmembrane receptors may also act as
14
331
transcriptional factors or modulators of expression (17, 20, 25, 34, 53, 62, 72). As T-cadherin
332
upregulates cyclin D1 (41) and may activate the serum response element (our unpublished
333
data), a similar function seems conceivable for T-cadherin.
334
We also found that T-cadherin is located in the centrosomes. A quantitative difference between
335
venous (HUVECs) and arterial (HPAECs) endothelial cells could be observed, as T-cadherin
336
was present in the centrosomes uniformly throughout the cell cycle (except telophase) in
337
HUVECs, but was most noticeable during metaphase in HPAECs. The mechanisms of protein
338
delivery to the centrosomes are poorly understood. For some proteins, such as RGS14 (13),
339
nuclear-cytoplasmic shuttling may be important. In this respect, it may be relevant that the two
340
lines of ECs studied here also differed in the proportion of nuclear T-cadherin. Since our data
341
suggest that T-cadherin may play a role in cytokinesis, it is also worth noting that some proteins
342
required for cytokinesis are sequestered into the nucleus during interphase (6). Centrosomal T-
343
cadherin may have a longer half life than T-cadherin in other compartments, since the
344
centrosomes were the last location where it could be detected upon siRNA-mediated depletion.
345
This is similar to the observations for centrosomal γ−tubulin (67). In line with these data, in HEK
346
293A cells (where T-cadherin expression is low), the endogenous T-cadherin could only be
347
observed in the centrosomes and to a minimal extent in the nucleus.
348
The role of T-cadherin in the centrosomes may be linked to the control of their duplication, since
349
siRNA-mediated T-cadherin depletion resulted in an increased fraction of cells with multiple
350
centrosomes. Since the data were collected for the cells that have an increased number of
351
centrosomes without nuclear duplication, this reflects a defect in centrosomal duplication per se.
352
Notably, depletion of most centrosomal proteins leads to cell cycle arrest and defects in
353
centrosome structure (52).
354
Our data also suggest that T-cadherin may play a role in cytokinesis, since it transiently
355
concentrates in the midbody during telophase, and its overexpression leads to an increased
15
356
proportion of cells with more than one nucleus. Notably, both the expression of T-cadherin (39)
357
and the proportion of bi- and multinucleated ECs (69) are elevated in atherosclerotic lesions.
358
Aneuploidy, which may be caused by different mitotic aberrations, including errors in
359
centrosome duplication or defective cytokinesis, is a characteristic feature of many cancerous
360
cells. At the same time, T-cadherin expression is known to be either down- or upregulated in
361
many cancer cell lines (e.g. (71) and references therein). In the light of our findings, it would be
362
interesting to examine whether T-cadherin expression is affected in tumors of endothelial origin,
363
which are also characterized by centrosome abnormalities (31). We did not detect statistically
364
significant differences in the percentage of multinuclear cells upon T-cadherin depletion.
365
However, since T-cadherin promotes cell cycle progression in vascular cells (38), T-cadherin
366
depletion may “freeze” the culture in the same state it was before siRNA transfection.
367
Overall, our findings reported here point to novel role(s) of T-cadherin in the nucleus and/or in
368
cell division. Although localization of T-cadherin described here is rather unexpected, its links to
369
the cell cycle have been reported previously in endothelial and other cell types: (i) T-cadherin
370
has been identified among cell cycle-related genes upregulated in G1 phase fibroblasts (37). (ii)
371
T-cadherin increases the expression of cyclin D1 in ECs (41). (iii) Ectopic expression of Cdh1
372
(which localizes dynamically to the nucleus during interphase and to the centrosomes during
373
metaphase and anaphase and is involved in the destruction of cell cycle regulators (75)),
374
stimulates T-cadherin degradation in PC12 cells (5). Our data, which indicate that T-cadherin is
375
under control of proteasome-dependent degradation in HUVECs and may be degraded in
376
telophase, are compatible with a role of Cdh1 in T-cadherin degradation in ECs as well. (iv)
377
Enforced expression of T-cadherin induces cell cycle arrest in hepatocellular carcinoma cells
378
(12). (v) Three kinases that act downstream of T-cadherin (41) and supposedly transduce T-
379
cadherin signals from the PM, are also implicated in the control of centrosomes and/or mitotic
380
spindles: (a) GSK-3β regulates localization of the γ-tubulin ring complex to the spindle poles and
16
381
thereby controls the formation of proper mitotic spindles (32). (b) Akt may localize to the
382
centrosomes in a manner dependent on its phosphorylation at specific sites (43), and its
383
reduced levels result in incomplete centrosome migration around the nucleus and in bent
384
misoriented mitotic spindles (10, 11). (c) ILK has recently been found to localize to mitotic
385
centrosomes and to be essential for spindle pole organization and mitosis (23).
386
Since our data suggest that either depletion or overexpression of T-cadherin results in defects in
387
cell division, findings reported here suggest a mechanistic insight into a previously described
388
correlation between either downregulation or upregulation of T-cadherin with cancers. Possible
389
contributions of diverse pathways to the function of T-cadherin in cell division will be the subject
390
of future studies.
391
392
ACKNOWLEGMENTS We would like to thank Drs Kalpana Ghoshal and Samson Jacob (Ohio
393
State University) for the FLAG-tagged T-cadherin constructs, Mr Aleksandar Krbanjevic for
394
plasmid purification, and Dr Jasmina Profirovic for critical reading of the manuscript.
395
396
17
397
REFERENCES
398
1.
Albi E and Viola Magni MP. The role of intranuclear lipids. Biol Cell 96: 657-667, 2004.
399
2.
Alonso-Nunez ML, An H, Martin-Cuadrado AB, Mehta S, Petit C, Sipiczki M, del Rey
400
F, Gould KL, and de Aldana CR. Ace2p controls the expression of genes required for cell
401
separation in Schizosaccharomyces pombe. Mol Biol Cell 16: 2003-2017, 2005.
402
3.
403
and transport of small molecules by caveolae. Science 255: 410-411, 1992.
404
4.
405
the mitochondria and affects mitochondrial morphology and motility. Faseb J 22: 2821-2831,
406
2008.
407
5.
408
factor induces proteasomal degradation of T-cadherin that requires tyrosine phosphorylation of
409
its cadherin domain. J Biol Chem 282: 27171-27180, 2007.
410
6.
411
860, 2007.
412
7.
413
and Pidard D. Proteolytic regulation of the urokinase receptor/CD87 on monocytic cells by
414
neutrophil elastase and cathepsin G. J Immunol 172: 540-549, 2004.
415
8.
416
of eukaryotic proteins using dipeptide composition and PSI-BLAST. Nucleic Acids Res 32:
417
W414-419, 2004.
418
9.
419
phosphatase 5 is dependent on the carboxy-terminal region. FEBS Lett 491: 279-284, 2001.
Anderson RG, Kamen BA, Rothberg KG, and Lacey SW. Potocytosis: sequestration
Andreeva AV, Kutuzov MA, and Voyno-Yasenetskaya TA. G alpha12 is targeted to
Bai S, Datta J, Jacob ST, and Ghoshal K. Treatment of PC12 cells with nerve growth
Barr FA and Gruneberg U. Cytokinesis: placing and making the final cut. Cell 131: 847-
Beaufort N, Leduc D, Rousselle JC, Magdolen V, Luther T, Namane A, Chignard M,
Bhasin M and Raghava GP. ESLpred: SVM-based method for subcellular localization
Borthwick EB, Zeke T, Prescott AR, and Cohen PT. Nuclear localization of protein
18
420
10.
421
regulates centrosome migration and spindle orientation in the early Drosophila melanogaster
422
embryo. J Cell Biol 180: 537-548, 2008.
423
11.
424
Cell Cycle 7: 2621-2625, 2008.
425
12.
426
cadherin in human hepatocellular carcinoma cells. Int J Cancer 123: 1043-1052, 2008.
427
13.
428
shuttling protein that traffics to promyelocytic leukemia nuclear bodies following heat shock. J
429
Biol Chem 280: 805-814, 2005.
430
14.
431
independent constitutive endocytosis and recycling of uPAR. PLoS ONE 3: e3730, 2008.
432
15.
433
the low adhesive capacity of human T-cadherin from the NMR structure of its N-terminal
434
extracellular domain. J Biol Chem, 2008.
435
16.
436
impairs centrosome-dependent microtubule nucleation and organization. Mol Biol Cell 19: 1220-
437
1229, 2008.
438
17.
439
Stachowiak MK. Fibroblast growth factor receptor-1 (FGFR1) nuclear dynamics reveal a novel
440
mechanism in transcription control. Mol Biol Cell 20: 2401-2412, 2009.
441
18.
442
neuronal death in the developing zebrafish. Dev Biol 321: 175-187, 2008.
443
19.
444
fistulas in rabbits. Circ Res 31: 546-556, 1972.
Buttrick GJ, Beaumont LM, Leitch J, Yau C, Hughes JR, and Wakefield JG. Akt
Buttrick GJ and Wakefield JG. PI3-K and GSK-3: Akt-ing together with microtubules.
Chan DW, Lee JM, Chan PC, and Ng IO. Genetic and epigenetic inactivation of T-
Cho H, Kim DU, and Kehrl JH. RGS14 is a centrosomal and nuclear cytoplasmic
Cortese K, Sahores M, Madsen CD, Tacchetti C, and Blasi F. Clathrin and LRP-1-
Dames SA, Bang E, Haussinger D, Ahrens T, Engel J, and Grzesiek S. Insights into
Didier C, Merdes A, Gairin JE, and Jabrane-Ferrat N. Inhibition of proteasome activity
Dunham-Ems SM, Lee YW, Stachowiak EK, Pudavar H, Claus P, Prasad PN, and
Emond MR and Jontes JD. Inhibition of protocadherin-alpha function results in
Fallon JT and Stehbens WE. Venous endothelium of experimental arteriovenous
19
445
20.
446
translocation of Flk-1/KDR, endothelial nitric oxide synthase, and caveolin-1 in vascular
447
endothelial cells. Biochem Biophys Res Commun 256: 192-197, 1999.
448
21.
449
Fujita Y. A role for the cleaved cytoplasmic domain of E-cadherin in the nucleus. J Biol Chem,
450
2008.
451
22.
452
phosphatidylinositol structures. Annu Rev Biochem 57: 285-320, 1988.
453
23.
454
kinase localizes to the centrosome and regulates mitotic spindle organization. J Cell Biol 180:
455
681-689, 2008.
456
24.
457
Resink TJ. Use of multicellular tumor spheroids to dissect endothelial cell-tumor cell
458
interactions: a role for T-cadherin in tumor angiogenesis. FEBS Lett 581: 4523-4528, 2007.
459
25.
460
MC. Endosomal transport of ErbB-2: mechanism for nuclear entry of the cell surface receptor.
461
Mol Cell Biol 25: 11005-11018, 2005.
462
26.
463
Marrache AM, Checchin D, Sennlaub F, Hou X, Nader M, Bkaily G, Ribeiro-da-Silva A,
464
Goetzl EJ, and Chemtob S. Modulation of pro-inflammatory gene expression by nuclear
465
lysophosphatidic acid receptor type-1. J Biol Chem 278: 38875-38883, 2003.
466
27.
467
zonula occludens-1, localizes to the nucleus before the maturation and during the remodeling of
468
cell-cell contacts. Proc Natl Acad Sci U S A 93: 10779-10784, 1996.
469
28.
470
Nucleic Acids Res 32: D122-124, 2004.
Feng Y, Venema VJ, Venema RC, Tsai N, and Caldwell RB. VEGF induces nuclear
Ferber EC, Kajita M, Wadlow A, Tobiansky L, Niessen C, Ariga H, Daniel J, and
Ferguson MA and Williams AF. Cell-surface anchoring of proteins via glycosyl-
Fielding AB, Dobreva I, McDonald PC, Foster LJ, and Dedhar S. Integrin-linked
Ghosh S, Joshi MB, Ivanov D, Feder-Mengus C, Spagnoli GC, Martin I, Erne P, and
Giri DK, Ali-Seyed M, Li LY, Lee DF, Ling P, Bartholomeusz G, Wang SC, and Hung
Gobeil F, Jr., Bernier SG, Vazquez-Tello A, Brault S, Beauchamp MH, Quiniou C,
Gottardi CJ, Arpin M, Fanning AS, and Louvard D. The junction-associated protein,
Guo T, Hua S, Ji X, and Sun Z. DBSubLoc: database of protein subcellular localization.
20
471
29.
472
nuclear function of gamma-protocadherins. J Biol Chem 280: 9313-9319, 2005.
473
30.
474
cadherin supports angiogenesis and adiponectin association with the vasculature in a mouse
475
mammary tumor model. Cancer Res 68: 1407-1416, 2008.
476
31.
477
chromosome and centrosome abnormalities. Cancer Res 65: 2507-2510, 2005.
478
32.
479
Hieda Y, and Higashiyama S. Membrane-anchored growth factor, HB-EGF, on the cell surface
480
targeted to the inner nuclear membrane. J Cell Biol 180: 763-769, 2008.
481
33.
482
vascular smooth muscle cell growth and migration signals. J Cell Biol 173: 417-429, 2006.
483
34.
484
ribosomal S6 kinase is a direct target for the nuclear fibroblast growth factor receptor 1 (FGFR1):
485
role in FGFR1 signaling. J Biol Chem 279: 29325-29335, 2004.
486
35.
487
regulation involves G2 phase arrest and requires p21(CIP1/WAF1) expression. Mol Cell Biol 23:
488
566-578, 2003.
489
36.
490
thrombosis. Angiology 7: 268-278, 1956.
491
37.
492
E2F in control of both DNA replication and mitotic functions as revealed from DNA microarray
493
analysis. Mol Cell Biol 21: 4684-4699, 2001.
494
38.
495
upregulation correlates with cell-cycle progression and promotes proliferation of vascular cells.
496
Cardiovasc Res 64: 132-143, 2004.
Haas IG, Frank M, Veron N, and Kemler R. Presenilin-dependent processing and
Hebbard LW, Garlatti M, Young LJ, Cardiff RD, Oshima RG, and Ranscht B. T-
Hida K and Klagsbrun M. A new perspective on tumor endothelial cells: unexpected
Hieda M, Isokane M, Koizumi M, Higashi C, Tachibana T, Shudou M, Taguchi T,
Hou R, Liu L, Anees S, Hiroyasu S, and Sibinga NE. The Fat1 cadherin integrates
Hu Y, Fang X, Dunham SM, Prada C, Stachowiak EK, and Stachowiak MK. 90-kDa
Huang ZY, Wu Y, Hedrick N, and Gutmann DH. T-cadherin-mediated cell growth
Impallomeni G. The alteration and regeneration of the endothelium in venous
Ishida S, Huang E, Zuzan H, Spang R, Leone G, West M, and Nevins JR. Role for
Ivanov D, Philippova M, Allenspach R, Erne P, and Resink T. T-cadherin
21
497
39.
498
V, Erne P, Resink T, and Tkachuk V. Expression of cell adhesion molecule T-cadherin in the
499
human vasculature. Histochem Cell Biol 115: 231-242, 2001.
500
40.
501
cadherin regulates vascular cell adhesion, phenotype and motility. Exp Cell Res 293: 207-218,
502
2004.
503
41.
504
an essential mediator for T-cadherin-dependent signaling via Akt and GSK3beta in endothelial
505
cells. Faseb J 21: 3083-3095, 2007.
506
42.
507
protects endothelial cells from oxidative stress-induced apoptosis. Faseb J 19: 1737-1739, 2005.
508
43.
509
involved in resumption of meiosis in mouse oocytes. Biol Cell 98: 111-123, 2006.
510
44.
511
Hancock JF, Mayor S, and Parton RG. Ultrastructural identification of uncoated caveolin-
512
independent early endocytic vehicles. J Cell Biol 168: 465-476, 2005.
513
45.
514
caveolae and non-caveolar lipid raft carriers. Biochim Biophys Acta 1745: 273-286, 2005.
515
46.
516
Cell Biol 15: 121-124, 2005.
517
47.
518
the nucleus and gene expression. Mol Biol Cell 18: 1064-1072, 2007.
519
48.
520
morphogenesis. J Cell Biol 169: 29-34, 2005.
Ivanov D, Philippova M, Antropova J, Gubaeva F, Iljinskaya O, Tararak E, Bochkov
Ivanov D, Philippova M, Tkachuk V, Erne P, and Resink T. Cell adhesion molecule T-
Joshi MB, Ivanov D, Philippova M, Erne P, and Resink TJ. Integrin-linked kinase is
Joshi MB, Philippova M, Ivanov D, Allenspach R, Erne P, and Resink TJ. T-cadherin
Kalous J, Solc P, Baran V, Kubelka M, Schultz RM, and Motlik J. PKB/AKT is
Kirkham M, Fujita A, Chadda R, Nixon SJ, Kurzchalia TV, Sharma DK, Pagano RE,
Kirkham M and Parton RG. Clathrin-independent endocytosis: new insights into
Kutay U and Guttinger S. Leucine-rich nuclear-export signals: born to be weak. Trends
Liao HJ and Carpenter G. Role of the Sec61 translocon in EGF receptor trafficking to
Luo Y and Radice GL. N-cadherin acts upstream of VE-cadherin in controlling vascular
22
521
49.
522
protocadherin Fat1 and translocation of its cytoplasmic domain to the nucleus. Exp Cell Res 307:
523
100-108, 2005.
524
50.
525
Philips MR. Rac1 accumulates in the nucleus during the G2 phase of the cell cycle and
526
promotes cell division. J Cell Biol 181: 485-496, 2008.
527
51.
528
Differential localization of Rho GTPases in live cells: regulation by hypervariable regions and
529
RhoGDI binding. J Cell Biol 152: 111-126, 2001.
530
52.
531
centrosome integrity induces p38-p53-p21-dependent G1-S arrest. Nat Cell Biol 9: 160-170,
532
2007.
533
53.
534
kinase receptors and associated proteins in the regulation of signaling and vascular function.
535
Circ Res 98: 743-756, 2006.
536
54.
537
of human T-cadherin binds lipoproteins. Biochem Biophys Res Commun 276: 1240-1247, 2000.
538
55.
539
aggresome-related process: a hypothesis. Lancet Neurol 3: 496-503, 2004.
540
56.
541
and Croce CM. Tcl1 enhances Akt kinase activity and mediates its nuclear translocation. Proc
542
Natl Acad Sci U S A 97: 3028-3033, 2000.
543
57.
544
Rac mediate endothelial cell polarization and detachment induced by T-cadherin. Faseb J 19:
545
588-590, 2005.
Magg T, Schreiner D, Solis GP, Bade EG, and Hofer HW. Processing of the human
Michaelson D, Abidi W, Guardavaccaro D, Zhou M, Ahearn I, Pagano M, and
Michaelson D, Silletti J, Murphy G, D'Eustachio P, Rush M, and Philips MR.
Mikule K, Delaval B, Kaldis P, Jurcyzk A, Hergert P, and Doxsey S. Loss of
Mukherjee S, Tessema M, and Wandinger-Ness A. Vesicular trafficking of tyrosine
Niermann T, Kern F, Erne P, and Resink T. The glycosyl phosphatidylinositol anchor
Olanow CW, Perl DP, DeMartino GN, and McNaught KS. Lewy-body formation is an
Pekarsky Y, Koval A, Hallas C, Bichi R, Tresini M, Malstrom S, Russo G, Tsichlis P,
Philippova M, Ivanov D, Allenspach R, Takuwa Y, Erne P, and Resink T. RhoA and
23
546
58.
547
V,
548
glycosylphosphatidylinositol- anchored T-cadherin on the surface of vascular endothelial cells:
549
role for Grp78/BiP in T-cadherin-dependent cell survival. Mol Cell Biol 28: 4004-4017, 2008.
550
59.
551
cadherin to the leading edge of migrating vascular cells in vitro: a function in vascular cell
552
motility? Histochem Cell Biol 120: 353-360, 2003.
553
60.
554
molecule in the nervous system lacks the conserved cytoplasmic region. Neuron 7: 391-402,
555
1991.
556
61.
557
George A. Endoplasmic reticulum chaperone protein GRP-78 mediates endocytosis of dentin
558
matrix protein 1. J Biol Chem 283: 29658-29670, 2008.
559
62.
560
internalization and nuclear translocation of fibroblast growth factor (FGF) receptor 1. DNA Cell
561
Biol 23: 538-548, 2004.
562
63.
563
Piatier-Tonneau D, Lemoine A, and Debuire B. Investigation in liver tissues and cell lines of
564
the transcription of 13 genes mapping to the 16q24 region that are frequently deleted in
565
hepatocellular carcinoma. Clin Cancer Res 8: 3178-3186, 2002.
566
64.
567
delivered to recycling endosomes via a distinct cdc42-regulated, clathrin-independent pinocytic
568
pathway. Dev Cell 2: 411-423, 2002.
569
65.
570
is a glycosylphosphatidylinositol-anchored membrane enzyme that provides a barrier to
571
endocytosis-mediated transfer of a foreign gene. J Biol Chem 282: 17132-17140, 2007.
Philippova M, Ivanov D, Joshi MB, Kyriakakis E, Rupp K, Afonyushkin T, Bochkov
Erne
P,
and
Resink
TJ.
Identification
of
proteins
associating
with
Philippova M, Ivanov D, Tkachuk V, Erne P, and Resink TJ. Polarisation of T-
Ranscht B and Dours-Zimmermann MT. T-cadherin, a novel cadherin cell adhesion
Ravindran S, Narayanan K, Eapen AS, Hao J, Ramachandran A, Blond S, and
Reilly JF, Mizukoshi E, and Maher PA. Ligand dependent and independent
Riou P, Saffroy R, Comoy J, Gross-Goupil M, Thiery JP, Emile JF, Azoulay D,
Sabharanjak S, Sharma P, Parton RG, and Mayor S. GPI-anchored proteins are
Shiokawa D, Matsushita T, Shika Y, Shimizu M, Maeda M, and Tanuma S. DNase X
24
572
66.
573
its soluble BMP cleavage product in neural crest delamination. Development 134: 491-501,
574
2007.
575
67.
576
tubulin at the midbody is required for the completion of cytokinesis during the mammalian cell
577
division. J Cell Sci 108 ( Pt 9): 2955-2962, 1995.
578
68.
579
31-39, 2000.
580
69.
581
morphometric indices of the endothelial nuclei of the aorta and pulmonary artery in
582
atherosclerosis. Arkh Patol 70: 37-41, 2008.
583
70.
584
Azmuko AA, Kashirina NM, Vlasik TN, Tkachuk VA, and Resink TJ. Identification of 130 kDa
585
cell surface LDL-binding protein from smooth muscle cells as a partially processed T-cadherin
586
precursor. Biochim Biophys Acta 1416: 155-160, 1999.
587
71.
588
Kondo T, Oga A, Kawauchi S, Hirabayashi K, Numa F, Ito T, Saito T, Sasaki K, and Hinoda
589
Y. Aneuploidy predicts outcome in patients with endometrial carcinoma and is related to lack of
590
CDH13 hypermethylation. Clin Cancer Res 14: 3354-3361, 2008.
591
72.
592
another age of discovery for cytokine action? Sci STKE 2007: pe69, 2007.
593
73.
594
research. Histol Histopathol 16: 1287-1293, 2001.
595
74.
596
endocytosis in endothelial cells proceed via syndecan-4-dependent activation of Rac1 and a
597
Cdc42-dependent macropinocytic pathway. J Cell Sci 117: 3189-3199, 2004.
Shoval I, Ludwig A, and Kalcheim C. Antagonistic roles of full-length N-cadherin and
Shu HB, Li Z, Palacios MJ, Li Q, and Joshi HC. A transient association of gamma-
Simons K and Toomre D. Lipid rafts and signal transduction. Nat Rev Mol Cell Biol 1:
Solovyeva NA LL, Lysenko, AI. Analysis of the heterogeneity of cytological and
Stambolsky DV, Kuzmenko YS, Philippova MP, Bochkov VN, Bespalova ZD,
Suehiro Y, Okada T, Okada T, Anno K, Okayama N, Ueno K, Hiura M, Nakamura M,
Swanson SM and Kopchick JJ. Nuclear localization of growth hormone receptor:
Takeuchi T and Ohtsuki Y. Recent progress in T-cadherin (CDH13, H-cadherin)
Tkachenko E, Lutgens E, Stan RV, and Simons M. Fibroblast growth factor 2
25
598
75.
599
related (fzr) increases natural killer cell-mediated cell death and suppresses tumor growth.
600
Blood 96: 259-263, 2000.
601
76.
602
Thomas PJ. Dynamic association of proteasomal machinery with the centrosome. J Cell Biol
603
145: 481-490, 1999.
604
77.
605
different microtubule-dependent processes. Mol Biol Cell 17: 2476-2487, 2006.
606
78.
607
regulator CDH1 and its regulation by phosphorylation. J Biol Chem 278: 12530-12536, 2003.
Wang CX, Fisk BC, Wadehra M, Su H, and Braun J. Overexpression of murine fizzy-
Wigley WC, Fabunmi RP, Lee MG, Marino CR, Muallem S, DeMartino GN, and
Zhou C, Cunningham L, Marcus AI, Li Y, and Kahn RA. Arl2 and Arl3 regulate
Zhou Y, Ching YP, Chun AC, and Jin DY. Nuclear localization of the cell cycle
608
609
610
26
611
FIGURE LEGENDS
612
Fig. 1. Endogenous T-cadherin is present in the nuclei in endothelial cells (EC). A: T-cadherin
613
domain architecture and the rabbit polyclonal T-cadherin antibodies used in this work.
614
Abbreviations: CD, cadherin domain; CRS, C-terminal recognition sequence; SP, signal peptide.
615
Antibodies (italicized) and positions of their epitopes are shown. The TK antibody
616
recognizes human but not mouse T-cadherin. The antibody designated as SC is from Santa
617
Cruz Biotechnology (cat. number sc-7940). B: Endogenous T-cadherin is present in the nuclei in
618
three lines of human ECs, derived from pulmonary artery (HPAEC), umbilical vein (HUVEC) and
619
microvasculature (HMVEC). Immunostaining was performed with T-cadherin antibodies (TK or
620
SC as indicated), and mouse monoclonal VE-cadherin antibody, followed by AlexaFluor 488-
621
and AlexaFluor 594-conjugated secondary antibodies, respectively. Nuclei were visualized with
622
DAPI (blue). Arrowheads and arrows indicate the presence of T-cadherin at the cell surface in
623
some cells and in a pair of punctate structures in a mitotic cell, respectively. Lower panel:
624
primary antibodies were omitted.
(70)
625
626
Fig. 2. Distribution of T-cadherin (105 kDa, mature glycosylated form; 130 kDa, partially
627
processed precursor) between nuclear and non-nuclear fractions of HPAECs. (A) and HUVECs
628
(B) and assessment of the purity of respective nuclear fractions (C). Untransfected cells or cells
629
transfected with FLAG-T-cadherin were fractionated into the nuclear (N, 1000 g pellet) and
630
soluble (S, 1000 g supernatant) fractions. Proteins were separated on SDS-PAGE, transferred
631
onto a PVDF membrane and probed with the TK or FLAG antibodies. Positions of molecular
632
weight markers (PageRuler, Fermentas) are indicated. In A and B, quantification of the
633
immunoblotting data for endogenous T-cadherin forms was performed using ImageJ software.
634
Data shown are means of three separate experiments (error bars, S.D.). H1, histone H1. C,
635
fractions shown in A and B were probed for various markers for indicated subcellular structures
27
636
and compartments (AJ; adherens junctions; ER, endoplasmic reticulum; PM, plasma membrane;
637
TJ, tight junctions). 1 Proteins that have been reported in the nucleus.
638
reported as functional partners of T-cadherin (see main text for references).
2
Proteins that have been
639
640
Fig. 3. T-cadherin is located in the centrosomes in HUVECs. Cells were immunostained with the
641
T-cadherin SC antibody and mouse monoclonal γ-tubulin antibody, followed by AlexaFluor 488-
642
and AlexaFluor 594-conjugated secondary antibodies, respectively. Nuclei were visualized with
643
DAPI (blue). The cell cycle phases are indicated (A: mitosis: prophase, metaphase, anaphase
644
and telophase; B: interphase: G1, S, G2). Centrosomes are indicated with arrows. In the
645
telophase image, the brightness in the green channel has been enhanced to visualize T-
646
cadherin in the midbody (*) and in centrosomes.
647
648
Fig. 4. T-cadherin is degraded in a proteasome-dependent manner in HUVECs (A), and co-
649
fractionates (B) and physically associates (C) with γ-tubulin. A: Confluent cells were incubated in
650
the presence of inhibitors (20 μM) or DMSO (control) as indicated, and T-cadherin levels
651
analyzed by Western blotting with the TK antibody. Right panel shows quantification of the
652
results (means of four replicates; error bars, S.D.). B: Centrosomes were isolated from
653
untransfected HUVECs as described under Materials and Methods. Sucrose gradient fractions
654
were analyzed for the presence of γ-tubulin (centrosomal marker) and T-cadherin. TL, total
655
lysate. C: HUVECs were transfected with the ΔC3-4 construct or empty vector. Cells from two
656
10 cm plates were subjected to immunoprecipitation with γ-tubulin or GFP (negative control)
657
antibodies as described under Materials and Methods. Immunoprecipitated material (IP) was
658
probed with the T-cadherin (SC) antibody. TL, total lysates.
659
28
660
Fig. 5. T-cadherin overexpression increases proportion of bi- or multinuclear cells. HPAECs
661
were transfected with the full length human T-cadherin construct. Cells were collected 24 h after
662
transfection, and T-cadherin expression analyzed by Western blotting (A) and by
663
immunofluorescence (B, upper and middle panels). Lower panel in B shows an example of
664
binuclear cells overexpressing T-cadherin 72 h after transfection. C: HUVECs transfected with
665
the T-cadherin construct or GFP were examined for alterations in morphology 72 h after
666
transfection. Percentage of cells with more than one nucleus is shown relative to the total cell
667
number counted (50-80 cells per replicate, 4 replicates; error bars, S.D.).
668
669
Fig. 6. siRNA-mediated depletion of T-cadherin in ECs results in multiple centrioles. A, B:
670
Downregulation of endogenous T-cadherin in HPAECs and HUVECs. Cells were transfected
671
with control (C) or T-cadherin (T) siRNA and analyzed by immunoblotting 24 or 72 h after
672
transfection (A) or by immunofluorescence 24 h after transfection (B; HPAECs). VE-cadherin
673
was examined as a specificity control. C: Multiple centrosomes in T cadherin-depleted HUVECs,
674
visualized with γ-tubulin antibody 72 h after transfection (methanol fixation). D: Z-sections (γ-
675
tubulin staining) of the cell shown in the bottom panel in (C). In C and D, centrosomes are
676
indicated by arrows. E and F: HUVECs transfected with T-cadherin or control siRNA were
677
examined for aberrations in the number of nuclei and centrosomes per cell, respectively, 72 h
678
after transfection. E: Percentage of cells with more than one nucleus is shown relative to the
679
total cell number counted (90-170 cells per replicate, 4 replicates; error bars, S.D.). F:
680
Percentage of cells with >2 centrosomes is shown relative to the total cell number counted;
681
interphase binuclear cells were disregarded (500 cells per replicate, 3 replicates; error bars,
682
S.D.).
683
29
684
Supplementary Fig. 1. A: FLAG-tagged mouse T-cadherin constructs used in this work
685
(designated according to Ref. (70)). For abbreviations, see Fig. 1. B-D:, Full-length and
686
truncated T-cadherin forms as detected by immunoblotting (B and D) and immunofluorescence
687
(C) using different antibodies. B: HUVECs were transfected with different T-cadherin constructs
688
as indicated, and their expression analyzed using FLAG antibody and two T-cadherin antibodies
689
(TK and SC, see Fig. 1). Note that the TK antibody does not recognize mouse T-cadherin, and
690
the SC antibody only recognizes the ΔC3-4 deletion mutant. In the TK panel, a short ECL
691
exposure is shown that gives signal intensity comparable to two other panels; endogenous T-
692
cadherin becomes visible at longer exposures (see Fig. 5A). Positions of molecular weight
693
markers are indicated. C: Expression of FLAG-tagged T-cadherin constructs in HUVEC,
694
visualized with SC (green) and FLAG (red) antibodies. Cells were collected 24 h after
695
transfection. D: Distribution of endogenous T-cadherin forms between nuclear and non-nuclear
696
fractions of HPAECs. Cells were fractionated as in Fig. 2, and T-cadherin detected using three
697
independently raised antibodies (see Fig. 1 for their epitopes). Positions of molecular weight
698
markers (Fermentas) are indicated. E: Effect of leptomycin B (LMB) treatment on distribution of
699
endogenous T-cadherin in HUVECs. Cells were treated with LMB (10 ng/ml) for 6 h and
700
fractionated into nuclear and non-nuclear fractions as in D. Relative distribution of 130 kDa and
701
105 kDa forms of T-cadherin was calculated separately from the ECL signals. Data shown are
702
means of 3 replicates (± S.D.).
703
704
Supplementary Fig. 2. Co-localization of T-cadherin with centrosomes. A: Z-sections showing
705
localization of T-cadherin (SC antibody) in a metaphase HPAEC from the experiment shown in
706
Fig. 1B. B: Co-localization of T-cadherin with γ-tubulin in HUVECs. The experiment was
707
performed as in Fig. 3, except that TK antibody was used for T-cadherin staining and para-
708
formaldehyde fixation was used in the left panel.
30
709
710
Supplementary Fig. 3. T-cadherin in HEK 293A cells. A: Endogenous T-cadherin is located
711
predominantly in the centrosomes. Untransfected HEK 293A cells were immunostained with T-
712
cadherin (SC) and γ-tubulin antibodies. B: An endogenous T-cadherin-positive 60 kDa protein
713
physically associates with γ-tubulin. HEK 293A cells from four 10 cm plates were subjected to
714
immunoprecipitation with the γ-tubulin or GFP (negative control) antibodies. Immunoprecipitated
715
material (IP) was probed with the T-cadherin (SC) antibody. TL, total lysate. C-E: HEK 293A
716
cells were transfected with the human T-cadherin construct or GFP and examined by Western
717
blotting (C) or immunostaining (D: an example of binuclear T-cadherin-expressing cells; nuclei
718
were visualized with DAPI), and were assessed for alterations in morphology 72 h after
719
transfection. Percentage of cells with more than one nucleus is shown in E relative to the total
720
cell number counted (150-200 cells per replicate, 3 replicates; error bars, S.D.).
721
31
CD1
EC1
SP
TK
CD2
SC
CD3
HUVEC
SC
CD4
CD5
EC5
C
SC
CRS
HMVEC
N
HPAEC
B
T-cadherin VE-cadherin
TK
HPAEC
A
HMVEC
(control)
Figure 1
SC
DAPI
Merge
HPAEC
WB: N
TK
S
kDa
130
95
H1
C
B
T-cadherin in nuclear
fraction (relative to total)
A
HPAEC
N S
TFLAG
WB: N S
TK
250
130
LAMP1
Rab5
Calreticulin
72
GRP782
ZO11
55
VE-cadherin1
35
Bcl2
Hsp901
95
27
17
11
N
S kDa
130
95
130
95
FLAG
H1
WB:
GM130
kDa
HUVEC
Akt11,2
Rac11,2
PP51
HPAEC HUVEC
N S N S
T-cadherin in nuclear
fraction (relative to total)
Figure 2
HUVEC
N S
Golgi
Lysosomes
Endosomes
ER
ER / PM
PM (TJ)
PM (AJ)
Mitochondria
Cytoplasm
kDa
250
130
95
72
55
35
27
17
Figure 3
Telophase Anaphase
Metaphase Prophase
A
T-cadherin J-Tubulin
Merge
B
G1
S
G2
*
T-cadherin J-Tubulin
Merge
Figure 4
A
T-cadherin levels
(relative to control)
2.5
2
1.5
1
0.5
0
Control I1
I2 MG
132
B
kDa
TL
1
2
3
4
5
6
7
55
8
9 10 WB:
J-Tub
130
105
TK
60
SC
C
WB:
SC
Vector T-cadherin
TL
IP:
J-Tub
GFP
Figure 5
A
T-cad
C
Pg: 0.1 0.3 0.5 0.5
WB:
T-cad
(TK)
Longer exposure
VE-cad
C
Cells with >1 nucleus
( % total )
Hsp90
12
*
10
8
6
4
2
0
GFP T-cad
B T-cadherin (TK)
VE-cadherin
Merge
Figure 6
C SC
J-Tubulin
Merge
VE-cad
Hsp90
8
6
4
2
0
siRNA: Control T-cad
VE-cadherin Merge
D
0.00 Pm
Z-sections (JJ-Tubulin)
T-cad siRNA Control siRNA
B
TK
E
Cells with >1 nucleus
( % total )
HUVEC
HPAEC
24 h
72 h
72 h
siRNA: C T
C T
C T
WB:
T-cad
0.66 Pm
0.22 Pm
0.88Pm
Pm
1.10
0.44 Pm
1.10 Pm
F
Mononuclear cells with
>2 centrosomes (% total)
A
2
1.5
1
0.5
0
1
2
siRNA: Control T-cad