ArticlePDF Available

Calumenin-15 facilitates filopodia formation by promoting TGF-?? superfamily cytokine GDF-15 transcription

Authors:

Abstract and Figures

Filopodia, which are actin-rich finger-like membrane protrusions, have an important role in cell migration and tumor metastasis. Here we identify 13 novel calumenin (Calu) isoforms (Calu 3-15) produced by alternative splicing, and find that Calu-15 promotes filopodia formation and cell migration. Calu-15 shuttles between the nucleus and cytoplasm through interacting with importin α, Ran GTPase, and Crm1. The phosphorylation of the threonine at position 73 (Thr-73) by casein kinase 2 (CK2) is essential for the nuclear import of Calu-15, and either Thr-73 mutation or inhibition of CK2 interrupts its nuclear localization. In the nucleus, Calu-15 increases the transcription of growth differentiation factor-15 (GDF-15), a member of the transforming growth factor-β (TGF-β) superfamily, via binding to its promoter region. Furthermore, Calu-15 induces filopodia formation mediated by GDF-15. Together, we identify that Calu-15, a novel isoform of Calu with phosphorylation-dependent nuclear localization, has a critical role in promoting filopodia formation and cell migration by upregulating the GDF-15 transcription.
Casein kinase 2 (CK2) phosphorylates calumenin (Calu)-15 at threonine at position 73 (Thr-73) and facilitates its nuclear localization. (a) The subcellular localization of Calu-15 wild-type (WT), T73A, or T73E. Left panels, representative pictures. Nuclear DNA was stained with DAPI (4',6-diamidino-2-phenylindole; blue). Bar, 10 μm. Right panel, the graph shows the percentage of cells with predominantly nuclear (N>C), both nuclear and cytoplasmic (N=C) or predominantly cytoplasmic (N<C) localization of enhanced green fluorescent protein (EGFP) fusion protein (n=3; >50 cells per experiment). Data are mean±S.E.M.; ***P<0.001; n.s., no significant difference (contingency table test for independence). (b) Lysates of HEK293T cells expressing Calu-15–2 × EGFP WT or T73A mutants (upper panel), and lysates of HEK293T cells expressing Calu-15–2 × EGFP treated with dimethylsulfoxide (DMSO) or 4,5,6,7-tetrabromo-2-azabenzimidazole (TBB; lower panel) were separated by Phos-tag SDS-polyacrylamide gel electrophoresis, then immunoblotted with antibody against EGFP. Arrows indicate the phosphorylated form. (c) HeLa cells transfected with Calu-15–2 × EGFP WT or T73E mutant were treated with DMSO or TBB for 10 h post transfection and then fixed for observation. Left panels, representative pictures. Nuclear DNA was stained with DAPI (blue). Bar, 10 μm. Right panel, the graphs show the percentage of cells with different localization patterns (n=3; >50 cells per experiment). Data are mean±S.E.M.; ***P<0.001; n.s., no significant difference (contingency table test for independence). (d) HeLa cells transfected with Calu-15–2 × EGFP WT, T73A, or T73E were treated with or without leptomycin B (LMB) before fixation and observation. Nuclear DNA was stained with DAPI (blue). Bar, 10 μm. The graphs show the percentage of cells with different localization patterns (n=3; >50 cells per experiment). Data are mean±S.E.M.; n.s., no significant difference (contingency table test for independence)
… 
Calumenin (Calu)-15 promotes the transcription of growth differentiation factor-15 (GDF-15). (a) Quantitative real-time PCR analysis of the relative mRNA expression levels of the indicated genes in the HeLa cells stably expressing Calu-15–EGFP (n=3). ABHD2, α-/β-hydrolase domain containing protein 2; NELL2, neural epidermal growth factor-like 2; ITPR1, inositol 1,4,5-triphosphate receptor, type 1; PML, promyelocytic leukemia; EGFP, as a control. Data are mean±S.E.M.; ***P<0.001 (unpaired two-tailed Student’s t-test). (b) Lysates of HEK293T cells expressing Calu-15–2 × EGFP or 2 × EGFP were immunoblotted (western blotting (WB)) with the indicated antibodies. Right panel, the graph shows the relative protein level of GDF-15 (n=4). Data are mean±S.E.M.; ***P<0.001 (unpaired two-tailed Student’s t-test). (c) Analysis of GDF-15 promoter activity by dual-luciferase reporter assay. The left panel shows the schematic structures of plasmids with different lengths of GDF-15 promoter followed by luciferase gene. The right graph indicates the relative luciferase activity in Calu-15–EGFP-overexpressing HeLa cells transfected with the indicated plasmids (n=4). EGFP, as a control. Data are mean±S.E.M.; **P<0.01, ***P<0.001 (unpaired two-tailed Student’s t-test). (d) Chromatin immunoprecipitation (ChIP) assay of HeLa cells expressing Calu-15–EGFP or EGFP. (e) Quantification of the GDF-15 promoter activity in HeLa cells expressing Calu-15–2 × EGFP wild type (WT), T73A, or T73E (n=3). Data are mean±S.E.M.; *P<0.05 (unpaired two-tailed Student’s t-test)
… 
Calumenin (Calu)-15 promotes cell migration and facilitates filopodia formation in a growth differentiation factor-15 (GDF-15)-dependent pathway. (a) Wound sizes of HeLa cells expressing Calu-15–EGFP or EGFP at the indicated time points (n=3). Data are mean±S.E.M.; ***P<0.001 (unpaired two-tailed Student’s t-test). (b) HeLa cells transfected with Calu-15–2 × EGFP or 2 × EGFP were stained with Atto 565 phalloidin to show the localization and distribution of filamentous (F)-actin. Left panel, representative pictures. Bar, 10 μm. Right panel, the graph shows the percentage of cells with filopodia (n=3; >100 cells per experiment). Data are mean±S.E.M.; ***P<0.001 (unpaired two-tailed Student’s t-test). (c) HeLa cells transfected with Calu-15–2 × EGFP wild type (WT), T73A, or T73E were stained with Atto 565 phalloidin. Left panel, representative pictures. Bar, 10 μm. Right panel, the graph shows the percentage of cells with filopodia (n=3; >100 cells per experiment). Data are mean±S.E.M.; ***P<0.001 (unpaired two-tailed Student’s t-test). (d) Conditioned medium collected from HEK293T cells transfected with Calu-15–2 × EGFP or 2 × EGFP were added into the cultures of HeLa cells, and then the HeLa cells were stained with Atto 565 phalloidin. Left panel, representative pictures. Bar, 10 μm. Right panel, the graph shows the percentage of cells with filopodia (n=3; >100 cells per experiment). Data are mean±S.E.M.; ***P<0.001 (unpaired two-tailed Student’s t-test). (e) At 48 h after GDF-15 shRNA transfection, the protein level of GDF-15–EGFP in HEK293T cells were examined by immunoblotting (western blotting (WB)). N.C., negative control; 88, 91, and 92 indicate GDF-15 shRNA with different target sequences; Pool, a mixture of GDF-15 shRNA 88, 91, and 92. (f) At 48 h after cotransfection with Calu-15–2 × EGFP and GDF-15 shRNA pool (Pool, lower panels) or N.C. shRNA (upper panels), HeLa cells were stained with Atto 565 phalloidin. Left panel, representative pictures. Bar, 10 μm. Right panel, the graph shows the percentage of cells with filopodia (n=3; >100 cells per experiment). Data are mean±S.E.M.; ***P<0.001 (unpaired two-tailed Student’s t-test). (g) Quantitative real-time PCR analysis of the relative mRNA expression levels of Calu-15 and GDF-15 in SW480 and SW620 cancer cells (n=5). Data are mean±S.E.M.; ***P<0.001 (unpaired two-tailed Student’s t-test). For b, c, d and f, to quantify the number of cells with filopodia, cells were scored positive when presenting at least 10 filopodia ≥4 μm in length
… 
Content may be subject to copyright.
OPEN
Calumenin-15 facilitates filopodia formation by
promoting TGF-bsuperfamily cytokine GDF-15
transcription
H Feng
1,3
, L Chen
1,3
, Q Wang
1,3
, B Shen
1
, L Liu
1
, P Zheng
1
,SXu
1
, X Liu
1
, J Chen
1,2
and J Teng*
,1
Filopodia, which are actin-rich finger-like membrane protrusions, have an important role in cell migration and tumor metastasis.
Here we identify 13 novel calumenin (Calu) isoforms (Calu 3–15) produced by alternative splicing, and find that Calu-15 promotes
filopodia formation and cell migration. Calu-15 shuttles between the nucleus and cytoplasm through interacting with importin a,
Ran GTPase, and Crm1. The phosphorylation of the threonine at position 73 (Thr-73) by casein kinase 2 (CK2) is essential for the
nuclear import of Calu-15, and either Thr-73 mutation or inhibition of CK2 interrupts its nuclear localization. In the nucleus, Calu-15
increases the transcription of growth differentiation factor-15 (GDF-15), a member of the transforming growth factor-b(TGF-b)
superfamily, via binding to its promoter region. Furthermore, Calu-15 induces filopodia formation mediated by GDF-15. Together,
we identify that Calu-15, a novel isoform of Calu with phosphorylation-dependent nuclear localization, has a critical role in
promoting filopodia formation and cell migration by upregulating the GDF-15 transcription.
Cell Death and Disease (2013) 4, e870; doi:10.1038/cddis.2013.403; published online 17 October 2013
Subject Category: Cancer
Filopodia, which are finger-like projections supported by
tightly parallel-bundled filamentous actin, are involved in
many key physiological and pathological processes,
1–3
whereas abundant filopodia are considered strongly correlated
with the enhanced cell migration and tumor metastasis.
3–5
Filopodia contain receptors for various extracellular signals,
including cytokines and growth factors, to sense the cell’s
surroundings during cell migration, then trigger the formation
of initial adhesion sites, the recruitment of focal adhesion
components, and finally the reorganization of the actin
network.
3,6–8
Transforming growth factor-b(TGF-b),
a cytokine, has been reported to induce the expression of
paxillin, a focal adhesion complex component, and fascin, an
actin filament-bundling protein, which in turn promotes
filopodia formation and cell migration.
4,7
Therefore, TGF-b
and fascin emerge as potential targets for cancer therapies.
4,9
Growth differentiation factor-15 (GDF-15) is a member of
the TGF-bsuperfamily proteins.
10,11
It is also known as
macrophage inhibitory cytokine-1, prostate-derived factor,
placental bone morphogenetic protein, non-steroidal
anti-inflammatory drug-activated gene-1, and placental
TGF-b.
10,11
Similar to other TGF-bsuperfamily members,
GDF-15 is first synthesized as a pro-protein, and then cleaved
and secreted in its active mature form.
12
It has been reported
to have roles in kinds of cellular processes such as cell
proliferation, migration, differentiation, and apoptosis.
13–17
GDF-15 has both anti- and pro-tumorigenic functions
according to different cell types and different developmental
stages in the tumor.
13,17,18
A high level of GDF-15 in serum is
associated with a poor patient survival in colorectal and
prostate carcinoma,
19,20
and the level of GDF-15 has also
been reported to increase during the transition of colonic
lesions to cancer initiation.
19
However, there is also evidence
that overexpression of GDF-15 induces the apoptosis of
breast cancer cells and inhibits the tumorigenicity of LN-Z308
glioblastoma cell line.
21,22
Therefore, to unravel the molecular
mechanisms that regulate GDF-15 is the key point for the
understanding and treatment of malignant tumor.
Calumenin (Calu) belongs to the CREC protein family,
which is composed of Cab45, reticulocalbin-1, reticulocalbin-2
(also known as ERC-55), reticulocalbin-3, and Calu.
23,24
These proteins all contain multiple EF-hand domains and are
encoded respectively by five genes.
23,24
Interestingly, most of
these genes produce isoforms by alternative mRNA splicing
and these isoforms usually have different subcellular localiza-
tions and physiological functions.
23,24
The CALU gene has
been reported to produce two isoforms, Calu-1 and
Calu-2 (also known as crocalbin),
25,26
having equal length
1
State Key Laboratory of Bio-membrane and Membrane Bio-engineering, Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of
Life Sciences, Peking University, Beijing, China and
2
Center for Quantitative Biology, Peking University, Beijing, China
*Corresponding author: J Teng, State Key Laboratory of Bio-membrane and Membrane Bio-engineering, Key Laboratory of Cell Proliferation and Differentiation of the
Ministry of Education, College of Life Sciences, Peking University, 5 Yiheyuan Road, Haidian District, Beijing 100871, China. Tel: þ86-10-62767044;
Fax: þ86-10-62755786; E-mail: junlinteng@pku.edu.cn
3
These authors contributed equally to this work.
Received 18.7.13; revised 04.9.13; accepted 11.9.13; Edited by G Melino
Keywords: calumenin-15; isoform; phosphorylation; GDF-15; transcription; filopodia
Abbreviations: Calu, calumenin; ChIP, chromatin immunoprecipitation; CK2, casein kinase 2; EGR-1, early growth response protein 1; FAK, focal adhesion kinase;
GDF-15, growth differentiation factor-15; LMB, leptomycin B; NLS, nuclear localization signal; NES, nuclear export signal; TBB, 4,5,6,7-tetrabromo-2-azabenzimidazole;
TGF-b, transforming growth factor-b
Citation: Cell Death and Disease (2013) 4, e870; doi:10.1038/cddis.2013.403
&
2013 Macmillan Publishers Limited All rights reserved 2041-4889/13
www.nature.com/cddis
(315 amino acids (aa)) with only exons 3 and 4 exchanged.
23
Recently, standard Edman degradation assay revealed that
both Calu-1 and Calu-2 possess an N-terminal signal peptide
(19 aa),
27
which leads to their translocations into the ER or
Golgi lumen.
28
Functionally, Calu-1 and Calu-2 regulate
vitamin K-dependent g-carboxylation activity
29
and participate
in calcium cycling by interacting with ryanodine receptor-1 and
ER Ca
2þ
-ATPase SERCA2.
30,31
Here we set out to determine whether there are other Calu
isoforms and identify that the CALU gene produces 13 novel
isoforms (named Calu 3–15) by alternative splicing, with only
one of them, Calu-15, possessing nuclear localization. Calu-
15 shuttles between the nucleus and cytoplasm, and this
process is regulated by its phosphorylation. Functionally,
Calu-15 increases GDF-15 transcription level in the nucleus,
which in turn induces filopodia formation and promotes cell
migration.
Results
Identification of Calu isoforms and their subcellular
localizations. To search for more alternatively spliced
isoforms of the CALU gene, we designed a pair of primers
localized in the first and last exon (Supplementary Figure
S1a), and performed PCR analysis. Several bands were
amplified from the cDNA of HeLa cells (Supplementary
Figure S1b), and 13 more splicing variants were identified by
sequencing. We named these 13 novel variants Calu 3–15
(GenBank accession number: HM002604–HM002616;
Figure 1a).
Previous reports have shown that Calu-1 and Calu-2 have
eight exons with mRNA lengths of B3.4 kb.
25,26
When the
exon organization of these isoforms was compared, we found
that Calu-3 and Calu-4 possessed a novel exon (exon 2 in
Figure 1a), leading to an increase of mRNA length to B4.2 kb.
In addition, Calu-3 and Calu-4 had extra 8 aa at the
N-terminus compared with Calu-1 and Calu-2
(Supplementary Figure S1c). As the N-terminal 19 aa of
Calu-1 and Calu-2 are the signal peptide,
27,28
we examined
whether the extra 8 aa interrupted this signal. The fluores-
cence assay showed that Calu-3–EGFP and Calu-4–EGFP
also colocalized with GRIP1-mRFP, a Golgi apparatus
marker, similar to Calu-1–EGFP and Calu-2–EGFP
(Figure 1b). Furthermore, EGFP fusion with the N-terminal
27 aa (8 þ19 aa) of Calu-3 and Calu-4 also colocalized with
GRIP1-mRFP (Supplementary Figure S1d). These results
suggest that the N-terminal 27 aa of Calu-3 and Calu-4 still
function as a signal peptide.
We also determined the subcellular distributions of the
other identified isoforms by overexpressing their EGFP fusion
proteins in HeLa cells. We found that only Calu-15 lacked the
signal peptide and showed nuclear accumulation (Figure 1b).
Together, the CALU gene encodes 15 isoforms, and only
Calu-15 lacks the N-terminal signal peptide and shows
nuclear localization.
Calu-15 shuttles between the nucleus and cytoplasm,
mediated by importin-a, Ran GTPase, and Crm1. To
confirm that Calu-15 shows the nuclear localization, we
tagged it with 2 EGFP and found that Calu-15–2 EGFP
was also localized in the nucleus (Figure 2a). Consistently,
b-Gal–EGFP-tagged Calu-15 also localized in the nucleus,
whereas the control b-Gal–EGFP did not (Supplementary
Figure S2). Thus, Calu-15 is able to drag 2 EGFP and
b-Gal–EGFP into the nucleus.
To determine the subcellular localization of endogenous
Calu-15, we used the antibody against Calu-1 and Calu-2,
which could also recognize the overexpressed Calu-15–
EGFP similar to the EGFP antibody (Figure 2b). Using this
antibody, we analyzed the nuclear and cytoplasmic fraction
proteins extracted from HeLa cells by western blotting, and
found that Calu-1 and Calu-2 were in the tubulin-containing
cytoplasmic fraction, whereas Calu-15 was mainly detected in
the nuclear fraction along with the nuclear marker lamin
(Figure 2c).
After confirming that Calu-15 is localized in the nucleus, we
next investigated whether it was through the classical path-
way to enter the nucleus. We performed immunoprecipitation
experiments and found that Calu-15 was associated with
importin-a(Figure 2d), a protein participating in the nuclear
entry process.
32,33
Besides, immunoprecipitation showed that
Calu-15 also interacted with Ran GTPase (Figure 2d), which
has been reported to disassociate the importin–cargo com-
plex and be required for the nuclear import process.
33,34
Moreover, the cotransfection with RanQ69L, which is a
mutant defective in GTPase activity,
35
largely inhibited the
nuclear localization of Calu-15 (Figure 2e). Therefore, the
nuclear import process of Calu-15 is mediated by importin-a
and regulated by Ran GTPase.
Interestingly, Calu-15 also interacted with the exportin
protein Crm1 (Figure 2d), which is reported to mediate
nucleocytoplasmic transport.
33,36
Therefore, it is reasonable
to hypothesize that Calu-15 might be exported from the
nucleus through a Crm1-dependent pathway. To test this, we
used leptomycin B (LMB), a small molecule that interferes
Crm1–cargo binding,
37
to suppress the Crm1-dependent
nuclear export. In cells overexpressing Calu-15–2 EGFP
due to its high level of nuclear accumulation, LMB treatment
could not increase the nuclear accumulation of Calu-15
(Figure 2e). However, in the presence of RanQ69L, LMB
treatment obviously induced Calu-15 to be localized in the
nucleus (Figure 2e). Thus, Calu-15 can shuttle between the
nucleus and cytoplasm, and this is mediated by importin-a,
Ran GTPase, and Crm1.
The nuclear localization of Calu-15 depends on phos-
phorylation at Thr-73 by CK2. Given that phosphorylation
can regulate the nuclear-cytoplasmic shuttling of proteins,
38,39
we analyzed the amino acid sequence of Calu-15
with bioinformatics tools
40
and found that the threonine at
position 73 (Thr-73) might be phosphorylated by casein
kinase 2 (CK2). This finding prompted us to examine whether
this site had a role in the nuclear-cytoplasmic shuttling of
Calu-15. We mutated this amino acid to alanine (T73A) and
glutamic acid (T73E) to mimic the unphosphorylated and
phosphorylated state, respectively, and examined their
subcellular localizations. To quantify the localization pattern,
we classified it into three types: (1) cells with predominantly
nuclear localization of EGFP fusion protein (N4C);
(2) with both nuclear and cytoplasmic localization (N¼C);
Calumenin-15 facilitates filopodia formation
H Feng et al
2
Cell Death and Disease
and (3) with predominantly cytoplasmic localization
(NoC). Compared with the wild-type Calu-15 (52.2% of N4C),
Calu-15-T73A showed no nuclear localization (0% of N4C),
whereas no significant difference was observed between
Calu-15-T73E and wild type (Figure 3a, P¼1.41 10
33
and
P¼0.10, respectively). Thus, these results suggest that the
phosphorylation of Calu-15 at Thr-73 is related to its nuclear
localization.
To confirm that Calu-15 is phosphorylated at Thr-73, we
used SDS-PAGE with 50 mM Phos-tag acrylamide, in which
phosphorylated bands migrate slower.
41
An upshifted phos-
phorylated band of Calu-15 was detected in wild type,
whereas this band almost disappeared in Calu-15-T73A
mutant (Figure 3b). Next, to determine whether it is CK2 that
phosphorylates the Thr-73 of Calu-15 and affects its nuclear
localization, we used 4,5,6,7-tetrabromo-2-azabenzimidazole
(TBB) to specifically inhibit CK2 activity
42
immediately after
the transfection of Calu-15–2 EGFP. TBB treatment
decreased the phosphorylation level of Calu-15 (Figure 3b),
and the nuclear localization of Calu-15 was also significantly
interrupted (from 75.3% to 32.3% of N4C) after 10 h TBB
treatment (Figure 3c, P¼5.27 10
10
). However, TBB
treatment did not change the subcellular localization of
Calu-15-T73E mutant (Figure 3c, P¼0.78), suggesting that
Figure 1 Fifteen calumenin (Calu) isoforms and their subcellular localizations. (a) Schematic picture shows the exon organization of 15 Calu isoforms encoded by the
CALU gene. The number at the top of the picture is the exon number. Black regions represent coding sequence, white regions represent untranslated mRNA sequence.
(b) Subcellular localizations of the enhanced green fluorescent protein (EGFP) fusion proteins of Calu isoforms (green). GRIP1-mRFP (red) marks the Golgi apparatus.
Bar, 10mm
Calumenin-15 facilitates filopodia formation
H Feng et al
3
Cell Death and Disease
CK2 promotes the nuclear localization of Calu-15 through
phosphorylation at Thr-73. As the loss of phosphorylation at
Thr-73 led to no nuclear localization of Calu-15 (Figure 3a), we
then examined whether it was due to the decrease of the
nuclear import or an increase of the nuclear export process.
The treatment of the nuclear export inhibitor LMB could not
increase the nuclear localization of Calu-15-T73A (Figure 3d,
P¼0.84), suggesting that the reason why the mutant
Calu-15–T73A showed no nuclear localization is not
the increase of nuclear export. Together, we conclude that
the phosphorylation of Calu-15 at Thr-73 by CK2 is
indispensable for its nuclear localization.
Calu-15 promotes GDF-15 transcription. To unravel the
physiological function of Calu-15 in the nucleus, we
examined the transcription levels of a group of genes
(Figure 4a). Quantitative real-time PCR analysis showed
that GDF-15 mRNA level was significantly increased in the
HeLa cell line stably expressing Calu-15–EGFP (Figure 4a,
P¼6.85 10
6
, and Supplementary Figure S3). Corre-
spondingly, GDF-15 protein level was also increased when
overexpressing Calu-15–2 EGFP, which is revealed by
western blotting (Figure 4b, P¼0.0006).
To investigate whether Calu-15 changes the GDF-15
expression level through interacting with its promoter, we
performed dual-luciferase reporter assay and found that
under the control of the promoter of GDF-15 (1 kb) the relative
luciferase activity was largely increased in Calu-15–EGFP-
expressing cells (Figure 4c, P¼3.67 10
–3
). Further analysis
showed that only a 200-bp region of the GDF-15 promoter was
sufficient (Figure 4c, P¼1.13 10
–3
). Moreover, ChIP assay
confirmed the interaction between Calu-15 and the promoter
Figure 2 Calumenin (Calu)-15 shuttles between the nucleus and the cytoplasm, mediated by importin-a, Ran GTPase, and Crm1. (a) Subcellular localization of Calu-15–
2enhanced green fluorescent protein (EGFP) fusion protein (green). Nuclear DNA was stained with DAPI (4’,6-diamidino-2-phenylindole; blue). Bar, 10 mm. The upper
panel shows the domain architecture of Calu-15–2 EGFP. (b) Immunoprecipitation (IP) with antibody against EGFP were performed in the lysates of HEK293T cells
expressing Calu-15–EGFP, followed by immunoblotting (western blotting (WB)) with antibody against Calu-1/2 (calu). Antibody against EGFP (EGFP) as a positive control.
IgG as a negative control. (c) Immunoblottings (WB) of endogenous Calu-15 in the cytoplasmic and nuclear fraction from HeLa cells. Asterisks indicate the band of Calu-1/2 or
Calu-15. Tubulin and lamin, respectively, labels the cytoplasmic and nuclear fraction. (d) IP with antibody against EGFP was performed in the lysates of HEK293T cells
expressing Calu-15–EGFP or EGFP, followed by immunoblotting (WB) with the indicated antibodies. (e) HeLa cells cotransfected with Calu-15–2 EGFP and RanQ69L-
mRFP were treated with (right panels) or without (left panels) leptomycin B (LMB) for 3 h, then fixed for observation. Nuclear DNA was stained with DAPI (blue). Bar, 10 mm
Calumenin-15 facilitates filopodia formation
H Feng et al
4
Cell Death and Disease
Figure 3 Casein kinase 2 (CK2) phosphorylates calumenin (Calu)-15 at threonine at position 73 (Thr-73) and facilitates its nuclear localization. (a) The subcellular
localization of Calu-15 wild-type (WT), T73A, or T73E. Left panels, representative pictures. Nuclear DNA was stained with DAPI (4’,6-diamidino-2-phenylindole; blue). Bar,
10 mm. Right panel, the graph shows the percentage of cells with predominantly nuclear (N4C), both nuclear and cytoplasmic (N ¼C) or predominantly cytoplasmic (NoC)
localization of enhanced green fluorescent protein (EGFP) fusion protein (n¼3; 450 cells per experiment). Data are mean±S.E.M.; ***Po0.001; n.s., no significant
difference (contingency table test for independence). (b) Lysates of HEK293T cells expressing Calu-15–2 EGFP WT or T73A mutants (upper panel), and lysates of
HEK293T cells expressing Calu-15–2 EGFP treated with dimethylsulfoxide (DMSO) or 4,5,6,7-tetrabromo-2-azabenzimidazole (TBB; lower panel) were separated by Phos-
tag SDS-polyacrylamide gel electrophoresis, then immunoblotted with antibody against EGFP. Arrows indicate the phosphorylated form. (c) HeLa cells transfected with Calu-
15–2 EGFP WT or T73E mutant were treated with DMSO or TBB for 10 h post transfection and then fixed for observation. Left panels, representative pictures. Nuclear DNA
was stained with DAPI (blue). Bar, 10 mm. Right panel, the graphs show the percentage of cells with different localization patterns (n¼3; 450 cells per experiment). Data are
mean±S.E.M.; ***Po0.001; n.s., no significant difference (contingency table test for independence). (d) HeLa cells transfected with Calu-15–2 EGFP WT, T73A, or T73E
were treated with or without leptomycin B (LMB) before fixation and observation. Nuclear DNA was stained with DAPI (blue). Bar, 10 mm. The graphs show the percentage of
cells with different localization patterns (n¼3; 450 cells per experiment). Data are mean±S.E.M.; n.s., no significant difference (contingency table test for independence)
Calumenin-15 facilitates filopodia formation
H Feng et al
5
Cell Death and Disease
of GDF-15 (Figure 4d). To further verify our findings, we
showed that the non-phosphorylation mimic mutant T73A,
with cytoplasmic localization pattern, was insufficient to
increase the relative luciferase activity, which was under the
control of the 200-bp region of the GDF-15 promoter, whereas
both the wild-type and the phosphorylation mimic mutant
T73E could (Figure 4e, P¼0.0337). Together, these results
suggest that nuclear Calu-15 regulates the GDF-15 transcription
via binding to its promoter region.
Calu-15 facilitates filopodia formation through a GDF-15-
mediated pathway. Interestingly, we found that overexpres-
sion of Calu-15–EGFP promoted cell migration in the wound-
healing assay (Figure 5a and Supplementary Figure S4).
Besides, Calu-15 also significantly increased the number and
size of filopodia (Figure 5b, P¼9.59 10
–5
), which facilitates
cell migration through highly active actin rearrangement.
13
Accordingly, the phosphorylation mimic mutant T73E also
induced filopodia formation, whereas the unphosphorylation
mimic mutant T73A failed (Figure 5c, P¼1.46 10
–5
,
P¼5.03 10
6
and P¼3.84 10
5
, respectively). All
these observations are consistent with previous reports that
GDF-15 is involved in the regulation of cell migration.
13
Considering that GDF-15 is a cytokine and is secreted into
the extracellular space, we then tested the conditioned
medium from Calu-15–2 EGFP-expressing HEK293T cells
and the filopodia formation was also induced (Figure 5d,
P¼1.38 10
–4
). Moreover, we knocked down GDF-15 in
Calu-15–2 EGFP-overexpressing HeLa cells and found
that the depletion of GDF-15 inhibited the function of
Calu-15 on filopodia formation (Figure 5f, P¼9.20 10
–4
).
Collectively, these data suggest that Calu-15 can induce
filopodia formation through GDF-15, which in turn promotes
cell migration.
To further confirm the correlation among Calu-15,
GDF-15, and tumor metastatic potential, we performed
quantitative real-time PCR analysis in colorectal cancer cell
lines SW480 and SW620, which were from the same patient.
Compared with the SW480 primary clone, the SW620
that originated from the metastatic lymph node shows a
higher tumor metastatic potential.
43,44
Consistently, both
Calu-15 and GDF-15 mRNA levels were significantly
increased in SW620 cells (Figure 5g, P¼2.25 10
7
and
P¼5.13 10
14
, respectively), suggesting a significant
correlation among Calu-15, GDF-15, and tumor metastatic
potential.
Discussion
In this study, we found that the CALU gene encodes 13 novel
isoforms besides Calu-1 and Calu-2. Among them, only Calu-
15 shows nuclear localization. We demonstrate that Calu-15
is phosphorylated at Thr-73 by CK2, and then enters the
nucleus to promote the transcription of GDF-15 via binding to
its promoter, resulting in enhanced filopodia formation and cell
migration (Figure 6). Moreover, the entry of Calu-15 into the
nucleus is mediated by importin-aand Ran GTPase, whereas
its export from the nucleus is mediated by Crm1.
Figure 4 Calumenin (Calu)-15 promotes the transcription of growth differentiation factor-15 (GDF-15). (a) Quantitative real-time PCR analysis of the relative mRNA
expression levels of the indicated genes in the HeLa cells stably expressing Calu-15–EGFP (n¼3). ABHD2, a-/b-hydrolase domain containing protein 2; NELL2, neural
epidermal growth factor-like 2; ITPR1, inositol 1,4,5-triphosphate receptor, type 1; PML, promyelocytic leukemia; EGFP, as a control. Data are mean±S.E.M.; ***Po0.001
(unpaired two-tailed Student’s t-test). (b) Lysates of HEK293T cells expressing Calu-15–2 EGFP or 2 EGFP were immunoblotted (western blotting (WB)) with the
indicated antibodies. Right panel, the graph shows the relative protein level of GDF-15 (n¼4). Data are mean±S.E.M.; ***Po0.001 (unpaired two-tailed Student’s t-test).
(c) Analysis of GDF-15 promoter activity by dual-luciferase reporter assay. The left panel shows the schematic structures of plasmids with different lengths of GDF-15 promoter
followed by luciferase gene. The right graph indicates the relative luciferase activity in Calu-15–EGFP-overexpressing HeLa cells transfected with the indicated plasmids
(n¼4). EGFP, as a control. Data are mean±S.E.M.; **Po0.01, ***Po0.001 (unpaired two-tailed Student’s t-test). (d) Chromatin immunoprecipitation (ChIP) assay of HeLa
cells expressing Calu-15–EGFP or EGFP. (e) Quantification of the GDF-15 promoter activity in HeLa cells expressing Calu-15–2 EGFP wild type (WT), T73A, or T73E
(n¼3). Data are mean±S.E.M.; *Po0.05 (unpaired two-tailed Student’s t-test)
Calumenin-15 facilitates filopodia formation
H Feng et al
6
Cell Death and Disease
One gene can produce more than one isoform to function
either similarly or differently.
23
Our results show that the CALU
gene produces 15 isoforms in human cells by alternative
splicing, while 13 of them are first identified. Among these 15
isoforms, Calu 1–14 all possess an N-terminal signal peptide
and might localize in the lumen of the endomembrane system.
However, only Calu-15, lacking the N-terminal 151 aa,
shows nuclear accumulation and has novel functions as a
transcription regulator. Interestingly, the homologue gene of
Drosophila melanogaster,SCF, also encodes a nuclear
isoform DmSCF (DNA supercoiling factor) through alternative
splicing.
45
Therefore, it suggests that the production of a
nuclear isoform of the CALU gene is evolutionarily conserved
and has a significant physiological function.
In eukaryotic cells, nuclear import receptors such as
importin-aand -brecognize the nuclear localization signal
Figure 5 Calumenin (Calu)-15 promotes cell migration and facilitates filopodia formation in a growth differentiation factor-15 (GDF-15)-dependent pathway. (a) Wound
sizes of HeLa cells expressing Calu-15–EGFP or EGFP at the indicated time points (n¼3). Data are mean±S.E.M.; ***Po0.001 (unpaired two-tailed Student’s t-test).
(b) HeLa cells transfected with Calu-15–2EGFP or 2 EGFP were stained with Atto 565 phalloidinto show the localization and distribution of filamentous (F)-actin. Left panel,
representative pictures. Bar, 10 mm. Right panel, the graph shows the percentage of cells with filopodia (n¼3; 4100 cells per experiment). Data are mean±S.E.M.;
***Po0.001 (unpaired two-tailed Student’s t-test). (c) HeLa cells transfected with Calu-15–2 EGFP wild type (WT), T73A, or T73E were stained with Atto 565 phalloidin. Left
panel, representative pictures. Bar, 10 mm. Right panel, the graph shows the percentage of cells with filopodia (n¼3; 4100 cells per experiment). Data are mean±S.E.M.;
***Po0.001 (unpaired two-tailed Student’s t-test). (d) Conditioned medium collected from HEK293T cells transfected with Calu-15–2 EGFP or 2 EGFP were added into
the cultures of HeLa cells, and then the HeLa cells were stained with Atto 565 phalloidin. Left panel, representative pictures. Bar, 10 mm. Right panel, the graph shows the
percentage of cells with filopodia (n¼3; 4100 cells per experiment). Data are mean±S.E.M.; ***Po0.001 (unpaired two-tailed Student’s t-test). (e) At 48 h after GDF-15
shRNA transfection, the protein level of GDF-15–EGFP in HEK293T cells were examined by immunoblotting (western blotting (WB)). N.C., negative control; 88, 91, and 92
indicate GDF-15 shRNA with different target sequences; Pool, a mixture of GDF-15 shRNA 88, 91, and 92. (f) At 48 h after cotransfection with Calu-15–2 EGFP and GDF-15
shRNA pool (Pool, lower panels) or N.C. shRNA (upper panels), HeLa cells were stained with Atto 565 phalloidin. Left panel, representative pictures. Bar, 10 mm. Right panel,
the graph shows the percentage of cells with filopodia (n¼3; 4100 cells per experiment). Data are mean±S.E.M.; ***Po0.001 (unpaired two-tailed Student’s t-test).
(g) Quantitative real-time PCR analysis of the relative mRNA expression levels of Calu-15 and GDF-15 in SW480 and SW620 cancer cells (n¼5). Data are mean±S.E.M.;
***Po0.001 (unpaired two-tailed Student’s t-test). For b,c,dand f, to quantify the number of cells with filopodia, cells were scored positive when presenting at least 10
filopodia Z4mm in length
Calumenin-15 facilitates filopodia formation
H Feng et al
7
Cell Death and Disease
(NLS) of the proteins and then form an import complex with
them to enter the nucleus,
33
whereas exportins such as Crm1
recognize the nuclear export signal (NES) and form an export
complex to exit.
33,36
In our experiment, Calu-15 interacts with
both importin-aand Crm1. However, we have not identified
any classical NLS and NES in Calu-15, suggesting that there
may be other proteins to mediate its nuclear-cytoplasmic
shuttling, or that Calu-15 contains non-classical NLS and
NES. Besides, we show that the phosphorylation of Calu-15 at
Thr-73 by CK2 is indispensable for its nuclear localization,
which is similar to recent reports that nuclear-cytoplasmic
shuttling can be regulated by protein modification such as
phosphorylation and dephosphorylation.
38,39
CK2 has been
reported to regulate the nuclear export of S6K1 II,
46
but in our
study it acts as a regulator to facilitate the nuclear import of
Calu-15. However, we still do not know whether the
phosphorylation at Thr-73 facilitates the binding of Calu-15
directly to importin-aor to some other scaffold proteins. More
detailed mechanism needs further investigation.
GDF-15 functions as a cytokine, participating in various
cellular processes.
13–17
Meanwhile, it has pleiotropic func-
tions in cancer progression.
17
Because of its importance, the
transcription level of GDF-15 is under stringent control of
various regulatory pathways. Many transcription factors have
been reported to regulate its expression, such as p53, early
growth response protein 1 (EGR-1), Sp1, nuclear factor-kB,
poly (ADP-ribose) polymerase-1, and hypoxia-inducible
factor-1a.
14,47–51
Our results show that Calu-15, as a new
GDF-15 regulator, binds to the promoter region of GDF-15,
and our preliminary data suggests that other factors might be
required for this binding (data not shown). The binding region
of Calu-15 to GDF-15 we identified ranges from 168 to
þ32 bp around the transcription start site, in which it also
contains one p53- (at þ21 bp), three Sp1- (within the 133 to
þ41 bp), and two EGR-1- (within the 73 to 51 bp) binding
sites.
21,47,48
Whether Calu-15 functions coordinately with
them to promote the expression of GDF-15 remains unclear,
however, it is evident that this 200-bp region of GDF-15
promoter is crucial for its transcription. In addition, we do not
exclude the possibility that Calu-15 regulates the transcription
activities of other genes.
GDF-15 has been reported to promote filopodia formation
and metastasis of human prostate cancer cells through the
focal adhesion kinase (FAK)-RhoA signaling pathway.
13
Here we show that Calu-15 functionally promotes cell
migration by facilitating filopodia formation and this function
depends on GDF-15. Therefore, future research to investi-
gate whether Calu-15 also promotes filopodia formation
through FAK-RhoA signaling pathway could facilitate our
understanding of the detailed mechanism. Besides, the
function of GDF-15 in tumor progression is multiple but the
regulatory mechanism of GDF-15 in cancer cells is still
incompletely understood, whereas tumor cells with abundant
filopodia are identified to be more malignant and invasive
during tumor progression.
3–5
Considering the above two
facts, Calu-15, as a novel regulator of GDF-15, might have a
pro-tumorigenic role in cancer development and serve as a
new target for cancer therapy.
Materials and Methods
Cell lines and culture. HeLa, HEK293T, and SW480 cells were cultured at
37 1C with 5% CO
2
in Dulbecco’s modified Eagle’s medium (GIBCO BRL, Grand
Island, NY, USA) containing 10% heat-inactivated fetal bovine serum.
52
SW620
cells were cultured at 37 1C without CO
2
in Leibovitz’s L-15 medium (GIBCO BRL)
containing 10% heat-inactivated fetal bovine serum.
RNA isolation and cDNA cloning. Cells were homogenized in TRIzol
reagent (Invitrogen, Carlsbad, CA, USA) and the total RNA was isolated as
described in Molecular Cloning (third version). Subsequently, total RNA and
Superscript III Reverse Transcriptase (Invitrogen) were used to synthesize the
first-strand cDNA, according to the manufacturers’ instructions. The cDNAs
synthesized were then used for Calu cloning and quantitative real-time PCR
analysis. All PCR products were delivered for sequencing (Invitrogen).
Vector construction. To gain all the transcripts of Calu isoforms, specific
primers were synthesized and isoforms were cloned into the EcoRI and SalI site
of the pEGFP-N3 vector (Clontech Laboratories, Mountain View, CA, USA).
2EGFP plasmid was constructed by cloning EGFP into the KpnI and SmaI site
of the pEGFP-N3 vector. All the other mutants of Calu-15 were cloned into the
EcoRI and SalI site of the 2 EGFP vector. The promoter of GDF-15 was cloned
into the pGL3-Basic vector (Promega, Madison, WI, USA).
Antibodies. Mouse anti-calu polyclonal antibody was produced previously.
52
Rabbit anti-EGFP polyclonal antibody was produced in our lab and was used in
immunoblotting and immunoprecipitation. Rabbit anti-importin-a, rabbit anti-lamin-
A and -C polyclonal antibody, and mouse anti-Ran GTPase polyclonal antibody
were from Professor CM Zhang (Peking University). Mouse anti-a-tubulin
monoclonal antibody (DM1a) was from Sigma (St. Louis, MO, USA). Mouse
anti-GAPDH monoclonal antibody (ab8245) was from Abcam (Cambridge, UK).
Mouse anti-GFP monoclonal antibody (clone 1E4) was from MBL (Woburn, MA,
USA). Rabbit anti-GDF-15 monoclonal antibody (D2A3) was from Cell Signaling
Technology (Danvers, MA, USA).
Immunoblotting analysis. Immunoblotting analysis were preformed as
described.
52
Briefly, the proteins were transferred onto PVDF membranes
(Millipore, Billerica, MA, USA) in a semidry transfer cell (Bio-Rad, Hercules, CA,
USA) after gel separation by SDS-PAGE. Next, the membranes were blocked in
3–5% milk in TTBS for 30 min and probed with primary antibodies at 4 1C
Figure 6 Proposed working model of calumenin (Calu)-15. Calu-15 undergoes
phosphorylation at threonine at position 73 (Thr-73) by casein kinase 2 (CK2) and
then promotes its entry into the nucleus. In the nucleus, Calu-15 can activate the
transcription of growth differentiation factor-15 (GDF-15) via binding to its promoter
region, and then leads to the formation of filopodia, which in turn promotes cell
migration
Calumenin-15 facilitates filopodia formation
H Feng et al
8
Cell Death and Disease
overnight. HRP-conjugated goat anti-mouse or anti-rabbit IgG (Jackson
ImmunoResearch Laboratories, West Grove, PA, USA) were used as secondary
antibodies. To test the phosphorylation, SDS-PAGE with 50 mM Phos-tag
acrylamide AAL-107 (Wako Pure Chemical Industries, Osaka, Japan) was used.
Transfection, RNAi, and immunofluorescence. HeLa cells were
transiently transfected by jetPEI (Polyplus, Illkirch, France) at B60% confluence
following the manufacturer’s instructions. At 24 h post transfection, cells were
washed in PBS and then fixed with freshly prepared 4% PFA (paraformaldehyde)
in PBS for 15 min at 37 1C. Subsequently, cells were permeabilized with 0.1%
Triton X-100 in PBS buffer for another 15 min and blocked by 3% BSA in PBS for
30 min. Next, they were incubated with primary and secondary antibodies or Atto
565 phalloidin (Sigma), and observed under a fluorescence microscope (Olympus,
Shinjuku, Tokyo, Japan) as previously described.
53
For GDF-15 knockdown, cells
were transfected with plasmids expressing short hairpin RNA targeting GDF-15
(88: 50-GCTCCAGACCTATGATGACTT-30; 91: 50-CCGGATACTCACGCCAGAA
GT-30; and 92: 50-CTATGATGACTTGTTAGCCAA-30; Sigma), then they were
lysed for western blotting or fixed for immunofluorescence at 48 h post
transfection.
Immunoprecipitation assay. For the immunoprecipitation assay,
HEK293T cells transfected with Calu-15–EGFP were lysed in the buffer (20 mM
Tris-HCl, 150 mM NaCl, 0.1% NP-40, 1 mM DTT, 0.5mM EGTA, 10% glycerol,
1 mM MgCl
2
, pH 7.9) containing a protease inhibitor cocktail (Roche, Basel,
Switzerland). Next, the total cell lysates were incubated with anti-EGFP antibody at
41C overnight. The protein samples that combined with the Protein A Sepharose
(GE Healthcare, Uppsala, Sweden) were collected and subjected to western
blotting assay.
Nuclear and cytoplasmic extraction. HeLa cells were first incubated
with buffer 1 (20 mM HEPES, 5 mM CH
3
COOK, 0.5 mM MgCl
2
, 1 mM DTT, pH
7.8) on ice for 10 min and then homogenized by 25 strokes of a glass Dounce
homogenizer. The cytoplasmic fraction was gained by first centrifugation at
4000 r.p.m. for 5 min and second centrifugation of the supernatant at 20000 g
for 30 min. The fraction remaining from the first centrifugation was incubated with
buffer 2 (20 mM HEPES, 5 mM CH
3
COOK, 0.5 mM MgCl
2
, 1 mM DTT, 0.4 M
NaCl, pH 7.8) at 4 1C for 9 min and then centrifuged at 14 000 r.p.m. for 30 min to
gain the nuclear fraction. Nuclear and cytoplasmic fractions were then subjected to
western blotting.
Quantitative real-time PCR. The ABI 7300 Detection System (Applied
Biosystems, Foster City, CA, USA) was used to perform quantitative real-time
PCR using the SYBR Green PCR Master Mix (Applied Biosystems) as previously
described.
54,55
The primers used for quantitative real-time PCR were listed in
Supplementary Table S1. GAPDH served as a reference control and the 2
DDCT
method was used.
56
Dual-luciferase reporter assay. Different lengths of the promoter region
of GDF-15 were amplified from genomic DNA and cloned into the pGL3-Basic
vector. Next, 50 ng of the pGL3-Basic vectors containing different lengths of GDF-
15 promoter were transfected into HeLa cells, together with the indicated plasmids
and 50 ng of TK-Renilla receptor gene as an internal control. After 24 h, the
luciferase activity of total cell lysates was tested by the Dual-Luciferase Reporter
Assay System (Promega).
Chromatin immunoprecipitation assay. ChIP assay was performed as
described.
57
Briefly, cells were cross-linked by adding a final concentration of 1%
formaldehyde for 10 min at room temperature and cross-linking was stopped by
adding glycine to a final concentration of 125 mM. Cells were then washed with
1PBS, resuspended in cell lysis buffer (5 mM Pipes (KOH), 85 mM KCl, 0.5%
NP-40, pH 8.0), and centrifuged to get the nuclei. The nuclear fraction was then
resuspended in nuclear lysis buffer (50 mM Tris, 10 mM EDTA, 1% SDS, pH 8.1),
sonicated, and centrifuged to get the supernatant containing chromatin shorter
than 500 bp. The supernatant was diluted five-fold in ChIP dilution buffer (0.01%
SDS, 1.1% Triton X-100, 1.2 mM EDTA, 16.7 mM Tris, 167 mM NaCl, pH 8.1) and
incubated with anti-EGFP antibody at 4 1C overnight. Protein A Sepharose beads
(GE Healthcare) were used to collect the immune complex and then washed
consecutively for 3–5 min with 1 ml of each solution: low-salt wash buffer (0.1%
SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris, 150 mM NaCl, pH 8.1), high-salt
wash buffer (0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20mM Tris, 500 mM NaCl,
pH 8.1), and LiCl wash buffer (0.25 M LiCl, 1% NP-40, 1% deoxycholate, 1 mM
EDTA, 10 mM Tris, pH 8.0), twice in 1 TE buffer (10 mM Tris, 1 mM EDTA,
pH 8.0). The complexes were eluted by adding elution buffer (1% SDS, 0.1 M
NaHCO
3
). Next, the formaldehyde cross-linking in the elutant was reversed and
the DNA was purified by a Gel Extraction Kit (CWBIO, Beijing, China). The purified
DNA fraction was then analyzed by PCR using appropriate primers.
Wound-healing assay. HeLa cells stably expressing EGFP or Calu-15–
EGFP were planted on coverslips at the same density. A micropipette tip was used
to scratch a wound. The detached cells were removed and the remaining cells
were cultured at 37 1C. The wound sizes at the same site were recorded as the
indicated time.
Conflict of Interest
The authors declare no conflict of interest.
Acknowledgements. We thank Professor WG Zhu from the Peking University
for SW480 cell line, Professor CM Zhang from the Peking University for the rabbit
anti-importin-a, and rabbit anti-lamin-A and -C polyclonal antibody and mouse anti-
Ran GTPase polyclonal antibody, and Professor IC Bruce for reading the
manuscript. This work was supported by the National Natural Science Foundation of
China (31271424) and the Major State Basic Research Development Program of
China (973 Program; 2010CB833705).
1. Swaney KF, Huang CH, Devreotes PN. Eukaryotic chemotaxis: a network of signaling
pathways controls motility, directional sensing, and polarity. Annu Rev Biophys 2010; 39:
265–289.
2. Hall A. The cytoskeleton and cancer. Cancer Metastasis Rev 2009; 28: 5–14.
3. Mattila PK, Lappalainen P. Filopodia: molecular architecture and cellular functions.
Nat Rev Mol Cell Biol 2008; 9: 446–454.
4. Sun J, He H, Xiong Y, Lu S, Shen J, Cheng A et al. Fascin protein is critical for transforming
growth factor beta protein-induced invasion and filopodia formation in spindle-shaped
tumor cells. J Biol Chem 2011; 286: 38865–38875.
5. Vignjevic D, Schoumacher M, Gavert N, Janssen KP, Jih G, Lae M et al. Fascin, a novel
target of beta-catenin-TCF signaling, is expressed at the invasive front of human colon
cancer. Cancer Res 2007; 67: 6844–6853.
6. Lidke DS, Lidke KA, Rieger B, Jovin TM, Arndt-Jovin DJ. Reaching out for signals: filopodia
sense EGF and respond by directed retrograde transport of activated receptors. J Cell Biol
2005; 170: 619–626.
7. Han X, Stewart JE Jr., Bellis SL, Benveniste EN, Ding Q, Tachibana K et al. TGF-beta1 up-
regulates paxillin protein expression in malignant astrocytoma cells: requirement for a
fibronectin substrate. Oncogene 2001; 20: 7976–7986.
8. Giancotti FG, Ruoslahti E. Integrin signaling. Science 1999; 285: 1028–1032.
9. Akhurst RJ, Hata A. Targeting the TGFbeta signalling pathway in disease. Nat Rev Drug
Discov 2012; 11: 790–811.
10. Bootcov MR, Bauskin AR, Valenzuela SM, Moore AG, Bansal M, He XY et al. MIC-1, a
novel macrophage inhibitory cytokine, is a divergent member of the TGF-beta superfamily.
Proc Natl Acad Sci USA 1997; 94: 11514–11519.
11. Bottner M, Suter-Crazzolara C, Schober A, Unsicker K. Expression of a novel member of
the TGF-beta superfamily, growth/differentiation factor-15/macrophage-inhibiting cytokine-
1 (GDF-15/MIC-1) in adult rat tissues. Cell Tissue Res 1999; 297: 103–110.
12. Uchida K, Chaudhary LR, Sugimura Y, Adkisson HD, Hruska KA. Proprotein convertases
regulate activity of prostate epithelial cell differentiation markers and are modulated in
human prostate cancer cells. J Cell Biochem 2003; 88: 394–399.
13. Senapati S, Rachagani S, Chaudhary K, Johansson SL, Singh RK, Batra SK.
Overexpression of macrophage inhibitory cytokine-1 induces metastasis of human
prostate cancer cells through the FAK-RhoA signaling pathway. Oncogene 2010; 29:
1293–1302.
14. Tan M, Wang Y, Guan K, Y Sun. PTGF-beta, a type beta transforming growth factor
(TGF-beta) superfamily member, is a p53 target gene that inhibits tumor cell growth via
TGF-beta signaling pathway. Proc Natl Acad Sci USA 2000; 97: 109–114.
15. Roninson IB. Tumor cell senescence in cancer treatment. Cancer Res 2003; 63: 2705–2715.
16. Ichikawa T, Suenaga Y, Koda T, Ozaki T, Nakagawara A. TAp63-dependent induction of
growth differentiation factor 15 (GDF15) plays a critical role in the regulation of keratinocyte
differentiation. Oncogene 2008; 27: 409–420.
17. Mimeault M, Batra SK. Divergent molecular mechanisms underlying the pleiotropic
functions of macrophage inhibitory cytokine-1 in cancer. J Cell Physiol 2010; 224: 626–635.
18. Liu T, Bauskin AR, Zaunders J, Brown DA, Pankhurst S, Russell PJ et al. Macrophage
inhibitory cytokine 1 reduces cell adhesion and induces apoptosis in prostate cancer cells.
Cancer Res 2003; 63: 5034–5040.
Calumenin-15 facilitates filopodia formation
H Feng et al
9
Cell Death and Disease
19. Brown DA, Ward RL, Buckhaults P, Liu T, Romans KE, Hawkins NJ et al. MIC-1 serum
level and genotype: associations with progress and prognosis of colorectal carcinoma.
Clin Cancer Res 2003; 9: 2642–2650.
20. Brown DA, Lindmark F, Stattin P, Balter K, Adami HO, Zheng SL et al. Macrophage
inhibitory cytokine 1: a new prognostic marker in prostate cancer. Clin Cancer Res 2009;
15: 6658–6664.
21. Li PX, Wong J, Ayed A, Ngo D, Brade AM, Arrowsmith C et al. Placental transforming
growth factor-beta is a downstream mediator of the growth arrest and apoptotic response
of tumor cells to DNA damage and p53 overexpression. J Biol Chem 2000; 275:
20127–20135.
22. Albertoni M, Shaw PH, Nozaki M, Godard S, Tenan M, Hamou MF et al. Anoxia induces
macrophage inhibitory cytokine-1 (MIC-1) in glioblastoma cells independently of p53 and
HIF-1. Oncogene 2002; 21: 4212–4219.
23. Honore B. The rapidly expanding CREC protein family: members, localization, function,
and role in disease. Bio Essays 2009; 31: 262–277.
24. Honore B, Vorum H. The CREC family, a novel family of multiple EF-hand, low-affinity
Ca(2 þ)-binding proteins localised to the secretory pathway of mammalian cells. FEBS
Lett 2000; 466: 11–18.
25. Vorum H, Liu X, Madsen P, Rasmussen HH, Honore B. Molecular cloning of a cDNA
encoding human calumenin, expression in Escherichia coli and analysis of its
Ca2
þ
-binding activity. Biochim Biophys Acta 1998; 1386: 121–131.
26. Hseu MJ, Yen CH, Tzeng MC. Crocalbin: a new calcium-binding protein that is also a
binding protein for crotoxin, a neurotoxic phospholipase A2. FEBS Lett 1999; 445:
440–444.
27. Tsukumo Y, Tsukahara S, Saito S, Tsuruo T, Tomida A. A novel endoplasmic reticulum
export signal: proline at the þ2-position from the signal peptide cleavage site. J Biol Chem
2009; 284: 27500–27510.
28. Vorum H, Hager H, Christensen BM, Nielsen S, Honore B. Human calumenin localizes to
the secretory pathway and is secreted to the medium. Exp Cell Res 1999; 248: 473–481.
29. Wajih N, Sane DC, Hutson SM, Wallin R. The inhibitory effect of calumenin on the vitamin
K-dependent gamma-carboxylation system. Characterization of the system in normal and
warfarin-resistant rats. J Biol Chem 2004; 279: 25276–25283.
30. Sahoo SK, Kim T, Kang GB, Lee JG, Eom SH, Kim do H. Characterization of calumenin-
SERCA2 interaction in mouse cardiac sarcoplasmic reticulum. J Biol Chem 2009; 284:
31109–31121.
31. Jung DH, Mo SH, DH Kim. Calumenin a multiple EF-hands Ca2 þ-binding protein,
interacts with ryanodine receptor-1 in rabbit skeletal sarcoplasmic reticulum. Biochem
Biophys Res Commun 2006; 343: 34–42.
32. Conti E, Uy M, Leighton L, Blobe l G, Kuriyan J. Crystallographic analysis of the recognition
of a nuclear localization signal by the nuclear import factor karyopherin alpha. Cell 1998;
94: 193–204.
33. Cook A, Bono F, Jinek M, Conti E. Structural biology of nucleocytoplasmic transport. Annu
Rev Biochem 2007; 76: 647–671.
34. Stewart M. Molecular mechanism of the nuclear protein import cycle. Nat Rev Mol Cell Biol
2007; 8: 195–208.
35. Liu Q, Yu J, Zhuo X, Jiang Q, Zhang C. Pericentrin contains five NESs and an NLS
essential for its nucleocytoplasmic trafficking during the cell cycle. Cell Res 2010; 20:
948–962.
36. Fornerod M, Ohno M, Yoshida M, Mattaj IW. CRM1 is an export receptor for leucine-rich
nuclear export signals. Cell 1997; 90: 1051–1060.
37. Kudo N, Matsumori N, Taoka H, Fujiwara D, Schreiner EP, Wolff B et al. Leptomycin B
inactivates CRM1/exportin 1 by covalent modification at a cysteine residue in the central
conserved region. Proc Natl Acad Sci USA 1999; 96: 9112–9117.
38. Ma RY, Tong TH, Cheung AM, Tsang AC, Leung WY, Yao KM. Raf/MEK/MAPK signaling
stimulates the nuclear translocation and transactivating activity of FOXM1c. J Cell Sci
2005; 118(Pt 4): 795–806.
39. Lidke DS, Huang F, Post JN, Rieger B, Wilsbacher J, Thomas JL et al. ERK nuclear
translocation is dimerization-independent but controlled by the rate of phosphorylation.
J Biol Chem 2010; 285: 3092–3102.
40. Obenauer JC, Cantley LC, Yaffe MB. Scansite 2.0: Proteome-wide prediction of cell
signaling interactions using short sequence motifs. Nucleic Acids Res 2003; 31:
3635–3641.
41. Kinoshita E, Kinoshita-Kikuta E, Koike T. Separation and detection of large
phosphoproteins using Phos-tag SDS-PAGE. Nat Protoc 2009; 4: 1513–1521.
42. Sarno S, Reddy H, Meggio F, Ruzzene M, Davies SP, Donella-Deana A et al. Selectivity of
4,5,6,7-tetrabromobenzotriazole, an ATP site-directed inhibitor of protein kinase CK2
(’casein kinase-2’). FEBS Lett 2001; 496: 44–48.
43. Chen ML, Liang LS, Wang XK. miR-200c inhibits invasion and migration in human colon
cancer cells SW480/620 by targeting ZEB1. Clin Exp Metastasis 2012; 29: 457–469.
44. Yehezkel G, Cohen L, Kliger A, Manor E, Khalaila I. O-linked beta-N-acetylglucosaminyla-
tion (O-GlcNAcylation) in primary and metastatic colorectal cancer clones and effect of
N-acetyl-beta-D-glucosaminidase silencing on cell phenotype and transcriptome. J Biol
Chem 2012; 287: 28755–28769.
45. Kobayashi M, Aita N, Hayashi S, Okada K, Ohta T, Hirose S. DNA supercoiling factor
localizes to puffs on polytene chromosomes in Drosophila melanogaster. Mol Cell Biol
1998; 18: 6737–6744.
46. Panasyuk G, Nemazanyy I, Zhyvoloup A, Bretner M, Litchfield DW, Filonenko V et al.
Nuclear export of S6K1 II is regulated by protein kinase CK2 phosphorylation at Ser-17.
J Biol Chem 2006; 281: 31188–31201.
47. Baek SJ, Kim JS, Moore SM, Lee SH, Martinez J, Eling TE. Cyclooxygenase inhibitors
induce the expression of the tumor suppressor gene EGR-1, which results in the up-
regulation of NAG-1, an antitumorigenic protein. Mol Pharmacol 2005; 67: 356–364.
48. Baek SJ, Horowitz JM, Eling TE. Molecular cloning and characterization of human
nonsteroidal anti-inflammatory drug-activated gene promoter. Basal transcription is
mediated by Sp1 and Sp3. J Biol Chem 2001; 276: 33384–33392.
49. Shim M, Eling TE. Protein kinase C-dependent regulation of NAG-1/placental bone
morphogenic protein/MIC-1 expression in LNCaP prostate carcinoma cells. J Biol Chem
2005; 280: 18636–18642.
50. Frizzell KM, Gamble MJ, Berrocal JG, Zhang T, Krishnakumar R, Cen Y et al.
Global analysis of transcriptional regulation by poly(ADP-ribose) polymerase-1 and
poly(ADP-ribose) glycohydrolase in MCF-7 human breast cancer cells. J Biol Chem 2009;
284: 33926–33938.
51. Krieg AJ, Rankin EB, Chan D, Razorenova O, Fernandez S, Giaccia AJ. Regulation of the
histone demethylase JMJD1A by hypoxia-inducible factor 1 alpha enhances hypoxic gene
expression and tumor growth. Mol Cell Biol 2010; 30: 344–353.
52. Wang Q, Feng H, Zheng P, Shen B, Chen L, Liu L et al. The intracellular transport and
secretion of calumenin-1/2 in living cells. PLoS One 2012; 7: e35344.
53. Wang Q, Shen B, Zheng P, Feng H, Chen L, Zhang J et al. Silkworm coatomers and their
role in tube expansion of posterior silkgland. PLoS One 2010; 5: e13252.
54. Wang Q, Teng J, Shen B, Zhang W, Guo Y, Su X et al. Characterization of kinesin-like
proteins in silkworm posterior silk gland cells. Cell Res 2010; 20: 713–727.
55. Wang Q, Chen L, Shen B, Liu Y, Chen J, Teng J. The tau-like protein in silkworm
(Bombyx mori) induces microtubule bundle formation. Front Biosci (Elite Ed) 2012; 4:
998–1008.
56. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time
quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001; 25: 402–408.
57. Xie W, Ling T, Zhou Y, Feng W, Zhu Q, Stunnenberg HG et al. The chromatin remodeling
complex NuRD establishes the poised state of rRNA genes characterized by bivalent
histone modifications and altered nucleosome positions. Proc Natl Acad Sci USA 2012;
109: 8161–8166.
Cell Death and Disease is an open-access journal
published by Nature Publishing Group.Thisworkis
licensed under a Creative Commons Attribution-NonCommercial-
NoDerivs 3.0 Unported License. To view a copy of this license, visit
http://creativecommons.org/licenses/by-nc-nd/3.0/
Supplementary Information accompanies this paper on Cell Death and Disease website (http://www.nature.com/cddis)
Calumenin-15 facilitates filopodia formation
H Feng et al
10
Cell Death and Disease
... It is encoded from the 7q32 region of the human genome and translated into a 315 amino acid protein containing a common CREC signal sequence, a putative N-glycosylation region, 6~7 EF-hand motifs, and a carboxyl-terminal with an inefficient retention HDEF signal sequence [1]. Currently, a total of 15 CALU isoforms have been identified in human [2]. Unlike the other CREC family members, CALU proteins can only be transported via cellular secretory mechanisms, and most of the CALU isoforms (CALU [1][2][3][4][5][6][7][8][9][10][11][12][13][14] are detected in the endoplasmic reticulum (ER), Golgi apparatus and extracellular medium [3][4][5]. ...
... Currently, a total of 15 CALU isoforms have been identified in human [2]. Unlike the other CREC family members, CALU proteins can only be transported via cellular secretory mechanisms, and most of the CALU isoforms (CALU [1][2][3][4][5][6][7][8][9][10][11][12][13][14] are detected in the endoplasmic reticulum (ER), Golgi apparatus and extracellular medium [3][4][5]. However, CALU-15, which lacks a signal peptide sequence, can only be transported between cellular cytosol and nucleus [2]. ...
... Unlike the other CREC family members, CALU proteins can only be transported via cellular secretory mechanisms, and most of the CALU isoforms (CALU [1][2][3][4][5][6][7][8][9][10][11][12][13][14] are detected in the endoplasmic reticulum (ER), Golgi apparatus and extracellular medium [3][4][5]. However, CALU-15, which lacks a signal peptide sequence, can only be transported between cellular cytosol and nucleus [2]. ...
Article
Full-text available
Metastasis is known as a key step in cancer recurrence and could be stimulated by multiple factors. Calumenin (CALU) is one of these factors which has a direct impact on cancer metastasis and yet, its underlined mechanisms have not been completely elucidated. The current study was aimed to identify CALU co-expressed genes, their signaling pathways, and expression status within the human cancers. To this point, CALU associated genes were visualized using the Cytoscape plugin BisoGenet and annotated with the Enrichr web-based application. The list of CALU related diseases was retrieved using the DisGenNet, and cancer datasets were downloaded from The Cancer Genome Atlas (TCGA) and analyzed with the Cufflink software. ROC curve analysis was used to estimate the diagnostic accuracy of DEGs in each cancer, and the Kaplan–Meier survival analysis was performed to plot the overall survival of patients. The protein level of the signature biomarkers was measured in 40 biopsy specimens and matched adjacent normal tissues collected from CRC and lung cancer patients. Analysis of CALU co-expressed genes network in TCGA datasets indicated that the network is markedly altered in human colon (COAD) and lung (LUAD) cancers. Diagnostic accuracy estimation of differentially expressed genes showed that a gene panel consisted of CALU, AURKA, and MCM2 was able to successfully distinguish cancer tumors from healthy samples. Cancer cases with abnormal expression of the signature genes had a significantly lower survival rate than other patients. Additionally, comparison of CALU, AURKA, and MCM2 proteins between healthy samples, early and advanced tumors showed that the level of these proteins was increased through normal–carcinoma transition in both types of cancers. These data indicate that the interactions between CALU, AURKA, and MCM2 has a pivotal role in cancer development, and thereby needs to be explored in the future.
... On the other hand, calumenin and its fifteen protein isoforms of varying lengths, demonstrate complex intra-cellular localization patterns in the secretory pathway, cytoplasm and the nucleus [22,44]. It is currently not known, whether calumenin, like CFTR, might recycle through EEA1 containing endocytic vesicles as well. ...
... (2) Calumenin might be mobilized from the ER in response to intra-cellular Ca 2+ signaling, that has been found to be increased in cells expressing F508del-CFTR [59]. (3) Calumenin might undergo alternative splicing into various isoforms that might in turn translocate to the cytoplasm and nucleus [22,44]. Alternative splicing of XBP1 transcript mediated by inositol receptor endonuclease 1 (IRE1) has been shown to be triggered during the unfolded protein response triggered in response to misfolded proteins in the endoplasmic reticulum [60]. ...
... Alternative splicing of XBP1 transcript mediated by inositol receptor endonuclease 1 (IRE1) has been shown to be triggered during the unfolded protein response triggered in response to misfolded proteins in the endoplasmic reticulum [60]. Calumenin isoforms could also be potentially phosphorylated and translocated into the nucleus [44] where they might elicit changes in gene expression. Previously it has been shown that calreticulin, another calcium sensitive chaperone in the ER, can affect the protein levels of myocyte enhancer factor (MEF) 2C, a cardiac specific transcription factor involved in cardiac development [61]. ...
Article
Full-text available
The cystic fibrosis transmembrane regulator (CFTR) is a cyclic-AMP dependent chloride channel expressed at the apical surface of epithelial cells lining various organs such as the respiratory tract. Defective processing and functioning of this protein caused by mutations in the CFTR gene results in loss of ionic balance, defective mucus clearance, increased proliferation of biofilms and inflammation of human airways observed in cystic fibrosis (CF) patients. The process by which CFTR folds and matures under the influence of various chaperones in the secretory pathway remains incompletely understood. Recently, calumenin, a secretory protein, belonging to the CREC family of low affinity calcium binding proteins has been identified as a putative CFTR chaperone whose biophysical properties and functions remain uncharacterized. We compared hydropathy, instability, charge, unfoldability, disorder and aggregation propensity of calumenin and other CREC family members with CFTR associated chaperones and calcium binding proteins, wild-type and mutant CFTR proteins and intrinsically disordered proteins (IDPs). We observed that calumenin, along with other CREC proteins, was significantly more charged and less folded compared to CFTR associated chaperones. Moreover like IDPs, calumenin and other CREC proteins were found to be less hydrophobic and aggregation prone. Phylogenetic analysis revealed a close link between calumenin and other CREC proteins indicating how evolution might have shaped their similar biophysical properties. Experimentally, calumenin was observed to significantly reduce F508del-CFTR aggregation in a manner similar to AavLEA1, a well-characterized IDP. Fluorescence microscopy based imaging analysis also revealed altered trafficking of calumenin in bronchial cells expressing F508del-CFTR, indicating its direct role in the pathophysiology of CF. In conclusion, calumenin is characterized as a charged protein exhibiting close similarity with IDPs and is hypothesized to regulate F508del-CFTR folding by electrostatic effects. This work provides useful insights for designing optimized synthetic structural correctors of CFTR mutant proteins in the future.
... CALU, a calcium-binding protein localized in the endoplasmic reticulum (ER), is mainly involved in such ER functions as protein folding and sorting. Besides, CALU has recently been shown to influence cell mobility, migration, invasion, and metastasis during particular events, such as tumorigenesis, wound healing, immune response, and coagulation [16][17][18][19][20][21]. Several studies have explored the relationship between CALU expression and survival and yielded relatively consistent results. ...
... Under normal physiological conditions, CALU primarily participates in regulating Ca 2+ -dependent protein folding, sorting and maturation in the ER [31], Ca 2+ homeostasis [32,33], and muscle contraction/relaxation [34]. However, in tumor microenvironment, CALU was reported to play a critical role in promoting a series of malignant phenotypes including cancer cell survival [21], filopodia formation and cell migration [20], invasiveness [12], metastasis [15,35], cancer development [10], and resistance to chemotherapy [13]. So far, very little is known about the biological function of CALU in glioma. ...
Article
Full-text available
Background Calumenin (CALU) has been reported to be associated with invasiveness and metastasis in some malignancies. However, in glioma, the role of CALU remains unclear. Methods Clinical and transcriptome data of 998 glioma patients, including 301 from CGGA and 697 from TCGA dataset, were included. R language was used to perform statistical analyses. Results CALU expression was significantly upregulated in more malignant gliomas, including higher grade, IDH wildtype, mesenchymal, and classical subtype. Gene Ontology analysis revealed that CALU-correlated genes were mainly enriched in cell/biological adhesion, response to wounding, and extracellular matrix/structure organization, all of which were strongly correlated with the epithelial-mesenchymal transition (EMT) phenotype. GSEA further validated the profound involvement of CALU in EMT. Subsequent GSVA suggested that CALU was particularly correlated with three EMT signaling pathways, including TGFβ, PI3K/AKT, and hypoxia pathway. Furthermore, CALU played synergistically with EMT key markers, including N -cadherin, vimentin, snail, slug, and TWIST1. Survival and Cox regression analysis showed that higher CALU predicted worse survival, and the prognostic value was independent of WHO grade and age. Conclusions CALU was correlated with more malignant phenotypes in glioma. Moreover, CALU seemed to serve as a pro-EMT molecular target and could contribute to predict prognosis independently in glioma.
... We next sought to examine how Climp63 is regulated. Our laboratory has been studying the physiological roles of the proteins in the CREC protein family ( Chen et al., 2016;Feng et al., 2013; Wang et al., 2015), and we found that the luminal width of ER sheets in COS7 cells overexpressing Calu1 ( Figure S1B) was significantly narrower (30 nm compared with 50 nm in control) when analyzing the ER morphology using electron microscopy (Figures 2A and 2B). As Climp63 is the only reported protein to support ER luminal width, we determined to examine whether Climp63 interacts with Calu1. ...
... Furthermore, reciprocal immunoprecipitations confirmed that endogenous Calu1 associated with endogenous Climp63 (Figures 2D and 2E). This interaction was specific because Calu2, another major isoform of the CALU gene ( Feng et al., 2013), did not associate with Climp63 ( Figure 2F). In addition, immunoprecipitation assays between Calu1 and three other ER-shaping proteins, Kinectin (KTN1), p180, and Atlastin (Atl2), showed that Calu1 did not interact with any of these proteins ( Figure S1C), suggesting a specific association between Calu1 and Climp63. ...
Article
Full-text available
The ER is composed of distinct structures like tubules, matrices, and sheets, all of which are important for its various functions. However, how these distinct ER structures, especially the perinuclear ER sheets, are formed remains unclear. We report here that the ER membrane protein Climp63 and the ER luminal protein calumenin-1 (Calu1) collaboratively maintain ER sheet morphology. We show that the luminal length of Climp63 is positively correlated with the luminal width of ER sheets. Moreover, the lumen-only mutant of Climp63 dominant-negatively narrows the lumen of ER sheets, demonstrating that Climp63 acts as an ER luminal bridge. We also reveal that Calu1 specifically interacts with Climp63 and antagonizes Climp63 in terms of both ER sheet distribution and luminal width. Together, our data provide insight into how the structure of ER sheets is maintained and regulated.
... CALU localizes to the entire secretory pathway, including the ER, Golgi apparatus, and the extracellular matrix. It appears that extracellular CALU inhibits cell migration, whereas nuclear isoform calumenin-15 promotes filopodia formation and cell migration, which suggests that this protein exerts different functions when localized in different sites [33,34]. RCN3 has been reported to be associated with the maturation of alveolar epithelial type II (AECII) cells during alveogenesis [35]. ...
Article
Full-text available
Background: Keloid is a pathological skin scar formation with complex and unclear molecular pathology mechanism. Novel biomarkers and associated mechanisms are needed to improve current therapies. Objectives: To identify novel biomarkers and underlying pathological mechanisms of keloids. Methods: Six pairs of keloid scar tissues and corresponding normal skin tissues were quantitatively analyzed by a high-resolution label-free mass spectrometry-based proteomics approach. Differential protein expression data was further analyzed by a comprehensive bioinformatics approach to identify novel biomarkers and mechanistic pathways for keloid formation. Candidate biomarkers were validated experimentally. Results: In total, 1359 proteins were identified by proteomic analysis. Of these, 206 proteins exhibited a significant difference in expression between keloid scar and normal skin tissues. RCN3 and CALU were significantly upregulated in keloids. RCN1 and PDGFRL were uniquely expressed in keloids. Pathway analysis suggested that the XBP1-mediated unfolded protein response (UPR) pathway was involved in keloid formation. Moreover, a PDGFRL centric gene coexpression network was constructed to illustrate its function in skin. Conclusions and clinical relevance: Our study proposed four novel biomarkers and highlighted the role of XBP1-mediated UPR pathway in the pathology of keloids. It provided novel biological insights that contribute to develop novel therapeutic strategies for keloids. This article is protected by copyright. All rights reserved.
... In knockout cells, it has been demonstrated that CSNK2A1 controls the abundance of tropomyosin negatively (D'Amore et al., 2019). Furthermore, the cytokine GDF-15 that promotes cell migration is significantly twofold up-regulated at both time points (Figure 2), and its expression is indirectly controlled by CK2 (Feng et al., 2013). ...
Article
Full-text available
Clostridioides difficile is a major cause of nosocomial infection worldwide causing antibiotic-associated diarrhea and some cases are leading to pseudomembranous colitis. The main virulence factors are toxin A and toxin B. Hypervirulent strains of C. difficile are linked to higher mortality rates and most of these strains produce additionally the C. difficile binary toxin (CDT) that possesses two subunits, CDTa and CDTb. The latter is responsible for binding and transfer of CDTa into the cytoplasm of target cells; CDTa is an ADP ribosyltransferase catalyzing the modification of actin fibers that disturbs the actin vs microtubule balance and induces microtubule-based protrusions of the cell membrane increasing the adherence of C. difficile . The underlying mechanisms remain elusive. Thus, we performed a screening experiment using MS-based proteomics and phosphoproteomics techniques. Epithelial Hep-2 cells were treated with CDTa and CDTb in a multiplexed study for 4 and 8 h. Phosphopeptide enrichment was performed using affinity chromatography with TiO2 and Fe-NTA; for quantification, a TMT-based approach and DDA measurements were used. More than 4,300 proteins and 5,600 phosphosites were identified and quantified at all time points. Although only moderate changes were observed on proteome level, the phosphorylation level of nearly 1,100 phosphosites responded to toxin treatment. The data suggested that CSNK2A1 might act as an effector kinase after treatment with CDT. Additionally, we confirmed ADP-ribosylation on Arg-177 of actin and the kinetic of this modification for the first time.
... Concomitantly, actin-bundling proteins such as fascin, villin, and α-actinin, bundle actin filaments. VEGF-A [54], GDF-5 [55], EGF [56], HGF [57], TNFα [58] and TNFβ [51], leptin [59] Estrogen [60], TGFβ1 [61], endothelin-3 [52] IL-5 [62], VEGF-A [63], pro-NGF [64], thymosin α1 [65], hepatoma-derived growth factor (HDGF) [66], TGFβ1 [67], NaF [68], KGF [69], SDF-1α [70], exosomes [71], osteopontin [72] EGF [73], PDGF [74], TGFβ [75], VEGF [76], HGF [77,78], SDF-1 [79] ECM stimuli Fibronectin [80], fibrinogen [81] Fibronectin [80], fibrinogen [81] Fibronectin [82,83], ...
Article
Full-text available
Inorganic nanoparticles have emerged as an attractive theranostic tool applied to different pathologies such as cancer. However, the increment in inorganic nanoparticle application in biomedicine has prompted the scientific community to assess their potential toxicities, often preventing them from entering clinical settings. Cytoskeleton network and the related adhesomes nest are present in most cellular processes such as proliferation, migration, and cell death. The nanoparticle treatment can interfere with the cytoskeleton and adhesome dynamics, thus inflicting cellular damage. Therefore, it is crucial dissecting the molecular mechanisms involved in nanoparticle cytotoxicity. This review will briefly address the main characteristics of different adhesion structures and focus on the most relevant effects of inorganic nanoparticles with biomedical potential on cellular adhesome dynamics. Besides, the review put into perspective the use of inorganic nanoparticles for cytoskeleton targeting or study as a versatile tool. The dissection of the molecular mechanisms involved in the nanoparticle-driven interference of adhesome dynamics will facilitate the future development of nanotheranostics targeting cytoskeleton and adhesomes to tackle several diseases, such as cancer.
... AG1478 and cetuximab results in reduced cell migration by inhibiting actin dynamics 59 . Calumenin-15 has been reported to increase filopodia formation by targeting member of TGF-β superfamily 60 . Similarly, jasplakinolide (Jasp) also induces F-actin polymerization by activating ERK and AKT pathways 61 . ...
Article
Full-text available
The intracellular Tau aggregates are known to be associated with Alzheimer’s disease. The inhibition of Tau aggregation is an important strategy for screening of therapeutic molecules in Alzheimer's disease. Several classes of dyes possess a unique property of photo-excitation, which is applied as a therapeutic measure against numerous neurological dysfunctions. Rose Bengal is a Xanthene dye, which has been widely used as a photosensitizer in photodynamic therapy. The aim of this work was to study the protective role of Rose Bengal against Tau aggregation and cytoskeleton modulations. The aggregation inhibition and disaggregation potency of Rose Bengal and photo-excited Rose Bengal were observed by in-vitro fluorescence, circular dichroism, and electron microscopy. Rose Bengal and photo-excited Rose Bengal induce minimal cytotoxicity in neuronal cells. In our studies, we observed that Rose Bengal and photo-excited Rose Bengal modulate the cytoskeleton network of actin and tubulin. The immunofluorescence studies showed the increased filopodia structures after photo-excited Rose Bengal treatment. Furthermore, Rose Bengal treatment increases the connections between the cells. Rose Bengal and photo-excited Rose Bengal treatment-induced actin-rich podosome-like structures associated with cell membranes. The in-vivo studies on UAS E-14 Tau mutant Drosophila suggested that exposure to Rose Bengal and photo-excited Rose Bengal efficiency rescues the behavioural and memory deficit in flies. Thus, the overall results suggest that Rose Bengal could have a therapeutic potency against Tau aggregation.
... The outcome was in concordance with that in colon cancer [32]. Distinct from the extracellular effect of CALU-1/− 2 in downregulated cancers [31], CALU-15 facilitated the nuclear expression of nucleus GDF-15, which then mainly promoted cell migration and tumour metastasis [33]; our study confirmed that Fig. 6 MTDH and CALU partially reversed the function of miR-let-7b and miR-let-7c in vitro. a Western blotting was used to detect MTDH and CALU influenced by co-transfection of miR-let-7b/miR-let-7c and MTDH/CALU in HMVII and GAK; GAPDH is shown as a loading control. ...
Article
Full-text available
Background: Mucosal melanoma with poor prognosis is a common histopathologic subtype of melanoma among Chinese and other Asian peoples. Regulated microRNAs (miRNAs) have been reported as oncogenes or tumour suppressors in melanoma. However, the roles of specific miRNAs in mucosal melanoma remain largely unknown. Here, we aimed to assess the biological functions, molecular mechanisms and clinical potential of miR-let-7b and miR-let-7c in mucosal melanoma. Methods: The expression of miR-let-7b and miR-let-7c in mucosal melanoma was determined by quantitative polymerase chain reaction (qPCR). Cutoff scores for miR-let-7b and miR-let-7c expressions were calculated through receiver operating characteristic (ROC) curve analysis in 106 mucosal melanoma patients according to recurrence. Correlations of miR-let-7b and miR-let-7c expression with clinicopathological characteristics, disease-free survival (DFS) and clinical benefits after treatment were then statistically analysed. The biological functions and molecular mechanisms of miR-let-7b and miR-let-7c were studied in vitro and in vivo. Results: The expression of miR-let-7b and miR-let-7c was decreased in 94 cases (88.7%) and 89 cases (84.0%) of 106 mucosal melanoma patients compared with mucosal nevi. A correlation was observed between the expression of miR-let-7b, miR-let-7c and DFS after surgery. In addition, overexpression of miR-let-7b or miR-let-7c inhibited mucosal melanoma cell growth, migration, invasion and metastasis and induced cell apoptosis and cell cycle arrest in vitro and in vivo. Mechanistically, miR-let-7b and miR-let-7c directly targeted metadherin (MTDH) and calumenin (CALU) and suppressed phospho-ERK in mucosal melanoma cells. MTDH and CALU reversed the partial function of miR-let-7b and miR-let-7c in vitro. Furthermore, progression-free survival (PFS) of mucosal melanoma patients upon temozolomide-based and paclitaxel-based chemotherapy was related to miR-let-7b and miR-let-7c expression. Overexpression of miR-let-7b or miR-let-7c in patient-derived xenograft (PDX) models and certain mucosal melanoma cells had better growth inhibition after temozolomide and paclitaxel treatment. MTDH reversed the sensitivity of miR-let-7b and miR-let-7c to paclitaxel in vitro. Conclusions: Our results suggested that miR-let-7b and miR-let-7c inhibited the recurrence of mucosal melanoma through inhibiting cell growth, migration, invasion and metastasis, inducing cell apoptosis and cell cycle arrest by targeting MTDH and CALU. In addition, miR-let-7b and miR-let-7c increased sensitivity to chemotherapeutic agents by targeting MTDH.
Chapter
Calumenin is a secretory pathway protein regulating different endoplasmic reticulum (ER) proteins such as the sarco-endoplasmic reticulum calcium ATPase (SERCA) pumps. Combined with its diverse cellular distribution, its calcium-binding ability, and its interaction with proteins involved in calcium signaling, it is easy to speculate on future description of important roles of calumenin in calcium homeostasis in many cell types, as it was initially observed in muscle cells. In this chapter, we describe basic techniques to modulate calumenin expression and detect its impact on ER calcium content using classic transfection and Western blot techniques, as well as ER calcium measurement using microplate reader.
Article
Full-text available
Many drugs that target transforming growth factor-β (TGFβ) signalling have been developed, some of which have reached Phase III clinical trials for a number of disease applications. Preclinical and clinical studies indicate the utility of these agents in fibrosis and oncology, particularly in augmentation of existing cancer therapies, such as radiation and chemotherapy, as well as in tumour vaccines. There are also reports of specialized applications, such as the reduction of vascular symptoms of Marfan syndrome. Here, we consider why the TGFβ signalling pathway is a drug target, the potential clinical applications of TGFβ inhibition, the issues arising with anti-TGFβ therapy and how these might be tackled using personalized approaches to dosing, monitoring of biomarkers as well as brief and/or localized drug-dosing regimens.
Article
Full-text available
The nutrient-sensing lipolytic enzyme adipose triglyceride lipase (ATGL) has a key role in adipose tissue function, and alterations in its activity have been implicated in many age-related metabolic disorders. In adipose tissue reduced blood vessel density is related to hypoxia state, cell death and inflammation. Here we demonstrate that adipocytes of poorly vascularized enlarged visceral adipose tissue (i.e. adipose tissue of old mice) suffer from limited nutrient delivery. In particular, nutrient starvation elicits increased activity of mitochondrial proline oxidase/dehydrogenase (POX/PRODH) that is causal in triggering a ROS-dependent induction of ATGL. We demonstrate that ATGL promotes the expression of genes related to mitochondrial oxidative metabolism (peroxisome proliferator-activated receptor-α, peroxisome proliferator-activated receptor-γ coactivator-1α), thus setting a metabolic switch towards fat utilization that supplies energy to starved adipocytes and prevents cell death, as well as adipose tissue inflammation. Taken together, these results identify ATGL as a stress resistance mediator in adipocytes, restraining visceral adipose tissue dysfunction typical of age-related metabolic disorders.Cell Death and Differentiation advance online publication, 4 October 2013; doi:10.1038/cdd.2013.137.
Article
Full-text available
Hormone-stimulated lipolysis is a rapid way to mobilize fat from its storage depot for use in peripheral tissues. By convention, activation of cytosolic lipases via the β-adrenergic receptor (ADRB2)-cAMP signaling pathway is the only molecular mechanism considered to liberate fatty acids from triglycerides stored in lipid droplets (LDs) of cells. Herein, we provide evidence that, aside from the activation of cytosolic lipases, autophagy contributes to this hormone-stimulated lipolysis. The ADRB2-stimulated lipolysis was reduced after inhibition of early or late autophagy using either pharmacological inhibitors or shRNA-mediated autophagic gene knockdown. ADRB2 stimulation has caused a marked increase in the autophagy-targeted LDs for lysosomal degradation, which is dependent on the LD-associated RAB7 as evidenced by the use of both shRNA-mediated RAB7 knockdown and a dominant-negative RAB7 mutant. In addition, RAB7 is involved in unstimulated (basal) lipolysis, and mediates the enhanced basal lipolysis in PLIN1/perilipin 1 knockdown fat cells. In conclusion, our results showed a contribution of lipophagy to both basal and hormone-stimulated lipolysis and that RAB7 plays a pivotal role in the regulation of this autolysosome-mediated lipid degradation in fat cells.
Article
Macrophage inhibitory cytokine-1 (MIC-1) is a divergent member of the tumor growth factor beta (TGF-beta) superfamily. Several observations suggest that it plays a role in colorectal carcinoma (CRC). In particular, MIC-1 is markedly up-regulated in colorectal cancers as well as in premalignant adenomas. This study examines the relationship of serum MIC-1 levels and genotypes to clinical and pathologic features of colonic neoplasia. Experimental Design: We confirmed the presence of MIC-1 in CRC tissue and the cell line CaCo-2. The normal range for serum MIC-1 levels was defined in 260 healthy blood donors, and the differences between normal subjects and 193 patients having adenomatous polyps or CRC were then determined. In a separate cohort of 224 patients, we evaluated the relationship of MIC-1 serum level and genotype to standard tumor parameters and outcome measures. MIC-1 was expressed in CRC tissue and the cancer cell line CaCo-2. There was a progressive increase in serum MIC-1 levels between normal individuals [mean (M) = 495 pg/ml, SD = 210), those with adenomatous polyps (M = 681 pg/ml, SD = 410), and those with CRC (M = 783 pg/ml, SD = 491)]. Serum MIC-1 level was correlated with the extent of disease so that the levels were higher in patients with higher Tumor-Node-Metastasis stage. There were significant differences in time to relapse and overall survival between subjects with different MIC-1 levels and genotypes. This study identifies a strong association between MIC-1 serum levels and neoplastic progression within the large bowel. We suggest that the measurement of serum MIC-1 levels and determination of MIC-1 genotype may have clinical use in the management of patients with CRC.
Article
Identification and characterization of p53 target genes would lead to a better understanding of p53 functions and p53-mediated signaling pathways. Two putative p53 binding sites were identified in the promoter of a gene encoding PTGF-, a type transforming growth factor (TGF-) superfamily member. Gel shift assay showed that p53 bound to both sites. Luciferase-coupled transactivation assay revealed that the gene promoter was activated in a p53 dose- as well as p53 binding site-dependent manner by wild-type p53 but not by several p53 mutants. The p53 binding and transactivationof the PTGF-beta promoter was enhanced by etoposide, a p53 activator, and was largely blocked by a dominant negative p53 mutant. Furthermore, expression of endogenous PTGF-beta was remarkably induced by etoposide in p53-positive, but not in p53-negative, cell lines. Finally, the conditioned medium collected from PTGF-beta-overexpressing cells, but not from the control cells, suppressed tumor cell growth. Growth suppression was not, however, seen in cells that lack functional TGF-beta receptors or Smad4, suggesting that PTGF-beta acts through the TGF-beta signaling pathway. Thus, PTGF-beta, a secretory protein, is a p53 target that could mediate p53-induced growth suppression in autocrinal as well as paracrinal fashions. The finding made a vertical connection between p53 and TGF-beta signaling pathways in controlling cell growth and implied a potential important role of p53 in inflammation regulation via PTGF-beta.
Article
By microsequencing and cDNA cloning we have identified the transformation-sensitive protein No. IEF SSP 9302 as the human homologue of calumenin. The nucleotide sequence predicts a 315 amino acid protein with high identity to murine and rat calumenin. The deduced protein contains a 19 amino acid N-terminal signal sequence, 7 EF-hand domains and, at the C-terminus, a HDEF sequence which has been reported to function as retrieval signal to the ER. The calumenin transcript is ubiquitously expressed in human tissue, at high levels in heart, placenta and skeletal muscle, at lower levels in lung, kidney and pancreas and at very low levels in brain and liver. Calumenin belongs to a family of multiple EF-hand proteins that include the ER localized proteins reticulocalbin and ERC-55 and the Golgi localized Cab45. Since its Ca2+ binding may be important for the function of the protein we have used microdialysis experiments in order to analyse for the affinity and the capacity of recombinant human (rh) calumenin. All 7 EF-hands of the protein are functional and bind Ca2+, each with an affinity of 1.6×103 M−1. The relatively low affinity for the EF-hands may suggest a role for the protein in Ca2+-dependent processes in the ER.
Article
The two most commonly used methods to analyze data from real-time, quantitative PCR experiments are absolute quantification and relative quantification. Absolute quantification determines the input copy number, usually by relating the PCR signal to a standard curve. Relative quantification relates the PCR signal of the target transcript in a treatment group to that of another sample such as an untreated control. The 2(-DeltaDeltaCr) method is a convenient way to analyze the relative changes in gene expression from real-time quantitative PCR experiments. The purpose of this report is to present the derivation, assumptions, and applications of the 2(-DeltaDeltaCr) method. In addition, we present the derivation and applications of two variations of the 2(-DeltaDeltaCr) method that may be useful in the analysis of real-time, quantitative PCR data. (C) 2001 Elsevier science.
Article
The mechanisms underlying the biological activity of metformin, a widely prescribed drug to treat type 2 diabetes, remain elusive. In a recent issue of Cell, Cabreiro et al. report that in C. elegans, metformin indirectly impacts lifespan by altering the methionine metabolism of its microbial partner E. coli (Cabreiro et al., 2013).
Article
Fascin, an actin-bundling protein overexpressed in all carcinomas, has been associated with poor prognosis, shorter survival, and more metastatic diseases. It is believed that fascin facilitates tumor metastasis by promoting the formation of invasive membrane protrusions. However, the mechanisms by which fascin is overexpressed in tumors are not clear. TGFβ is a cytokine secreted by tumor and mesenchymal cells and promotes metastasis in many late stage tumors. The pro-metastasis mechanisms of TGFβ remain to be fully elucidated. Here we demonstrated that TGFβ induced fascin expression in spindle-shaped tumor cells through the canonical Smad-dependent pathway. Fascin was critical for TGFβ-promoted filopodia formation, migration, and invasion in spindle tumor cells. More importantly, fascin expression significantly correlates with TGFβ1 and TGFβ receptor I levels in a cohort of primary breast tumor samples. Our results indicate that elevated TGFβ level in the tumor microenvironment may be responsible for fascin overexpression in some of the metastatic tumors. Our data also suggest that fascin could play a central role in TGFβ-promoted tumor metastasis.