ArticlePDF Available

Extracellular calumenin suppresses ERK1/2 signaling and cell migration by protecting fibulin-1 from MMP-13-mediated proteolysis

Authors:

Abstract and Figures

Extracellular proteins are vital for cell activities, such as cell migration. Calumenin is highly conserved among eukaryotes, but its functions are largely unclear. Here, we identify extracellular calumenin as a suppressor of cell migration and tumor metastasis. Calumenin binds to and stabilizes fibulin-1, leading to inactivation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) signaling. We further identify the minimal functional domain of calumenin (amino acids 74-138 and 214-280). Depletion of calumenin induces fibulin-1- and phospho-ERK1/2 (pERK1/2)-dependent promotion of cell migration. Consistently, in hepatocellular and pancreatic carcinoma, both calumenin and fibulin-1 are downregulated. Furthermore, we show that matrix metalloproteinase-13 (MMP-13) proteolyzes fibulin-1 and that calumenin protects fibulin-1 from cleavage by MMP-13. Calumenin, together with fibulin-1, also interacts with fibronectin and depends on both syndecan-4 and α5β1-integrin to suppress ERK1/2 signaling and inhibit cell migration. Thus, extracellular calumenin regulates fibulin-1 to have crucial roles in ERK1/2 signaling and cell migration.Oncogene advance online publication, 17 March 2014; doi:10.1038/onc.2014.52.
Extracellular calumenin suppresses cell migration. (a) Images of wound-healing assays of HeLa cells at 0, 24 and 48 h post-scratching, in the presence of conditioned medium containing HEK293T-cell-derived SP19-EGFP or calumenin-1/2-EGFP. EGFP-tagged protein in conditioned medium of HEK293T cells was detected by western blot using anti-EGFP antibody at 48 h post-transfection. (b) Quantification of wound sizes from (a) (n=8). (c) Transwell assays of HeLa cells, with HEK293T cells in the lower chamber transfected with SP19-EGFP or calumenin-1/2-EGFP. Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). EGFP-tagged protein in conditioned medium from HEK293T cells were detected by western blot using anti-EGFP antibody at 48 h post-transfection. (d) Invasion assays of HeLa cells with HEK293T cells in the lower chamber expressing IRES-EGFP or calumenin-1/2-IRES-EGFP. Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). Calumenin-1/2 in conditioned medium from HEK293T cells were detected by western blot at 48 h post-transfection. (e) Transwell assays of HeLa cells treated with eukaryotically expressed and purified calumenin-1-His-Strep (1 μg/ml). Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). (f) B16/F10 cells stably expressing IRES-EGFP or calumenin-1/2-IRES-EGFP were injected into C57BL/6J mice via the tail veins. Representative lungs and quantification of the numbers of metastatic pulmonary colonies 2 weeks post-injection are shown (n=12). (g) Transwell assays of HeLa cells treated with normal IgG or anti-calumenin-1/2 antibody (5 μg/ml). Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). (h) RNA interference efficiency of HeLa cells transfected with control or calumenin-1/2 siRNA for 72 h. HEK293T cells derived calumenin-1/2-containing conditioned medium was concentrated and added for rescue experiments. Calumenin-1/2 in total cell lysates and conditioned medium were detected by western blot. (i) Transwell assays of HeLa cells corresponding to (h). Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). (j) B16/F0 cells stably expressing control or calumenin-1/2 short hairpin RNA (shRNA) were injected into C57BL/6J mice via the tail veins. Representative lungs and quantification of the numbers of metastatic pulmonary colonies 3 weeks post-injection are shown (n=15 or 14). For a, c, d, e, g, i, scale bars, 100 μm; for f, j, scale bars, 1 cm. 293T, HEK293T cells; Calu, calumenin; CM, conditioned medium; Co.St., Coomassie blue staining; SP19, signal peptide (amino acids 1–19) of calumenin; TCL, total cell lysates. For b–e, g, i, data are mean±s.d.; for f, j, data are mean±s.e.m., *P<0.05, **P<0.01, ***P<0.001, determined by unpaired two-tailed Student’s t-test.
… 
Calumenin suppresses ERK1/2 phosphorylation and regulates actin organization. (a, b) Western blot of total cell lysates and conditioned medium from HeLa (a) or B16/F10 (b) cells overexpressing the indicated proteins for 48 h. Quantification of relative pERK1/2 to ERK1/2 levels (a) are shown (n=3). (c) Western blot of total cell lysates and conditioned medium from HeLa cells at 72 h post-transfection with control or calumenin-1/2 siRNA. HEK293T cells derived calumenin-1/2-containing conditioned medium was concentrated and added for rescue experiments. (d) Transwell assays of HeLa cells treated with MEK inhibitor U0126 (10 μM) and anti-calumenin-1/2 antibody (5 μg/ml). Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). Protein levels of total cell lysates from the treated HeLa cells were analyzed by western blot using anti-pERK1/2 and anti-calumenin antibodies. (e, f) pERK1/2 (e) and pMEK1/2 (f) of HeLa cells treated with eukaryotically expressed and purified calumenin-1-His-Strep (1 μg/ml) were detected by western blot. (g) pERK1/2 of HeLa cells overexpressing calumenin-1-EGFP or calumenin-1-Δ13/14-EGFP at 24 h post-transfection were detected by western blot. (h) Co-staining of paxillin and actin filaments in HeLa cells overexpressing SP19-EGFP or calumenin-1/2-EGFP for 72 h. (i, j) Co-staining of paxillin and actin filaments (i) or differential interference contrast images (j) of B10/F10 cells stably expressing IRES-EGFP or calumenin-1-IRES-EGFP. (k, l) Co-staining of paxillin and actin filaments (k) or differential interference contrast images (l) of B10/F0 cells stably expressing control short hairpin RNA (shRNA) or calumenin-1/2 shRNA. For h, i, k, scale bars, 10 μm; for j, l, scale bars, 100 μm. Calu, calumenin; CM, conditioned medium; Co.St., Coomassie blue staining; SP19, signal peptide (amino acids 1–19) of calumenin-1/2; TCL, total cell lysates. For a, d, data are mean±s.d., **P<0.01, ***P<0.001, determined by unpaired two-tailed Student’s t-test.
… 
Calumenin interacts with fibulin-1 in the extracellular space. (a) Immunoprecipitation assays with anti-EGFP antibody in conditioned medium and total cell lysates from HEK293T cells expressing calumenin-1-EGFP. Immunoprecipitates were subjected to sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS–PAGE) and Coomassie blue staining. Arrows indicate the proteins identified by liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis. (b) Conditioned medium from HEK293T cells expressing SP19-EGFP or calumenin-1/2-EGFP was immunoprecipitated with anti-EGFP antibody, and the immunoprecipitates were analyzed with anti-fibulin-1, anti-fibronectin and anti-EGFP antibodies by western blot. (c) Conditioned medium from HeLa cells was immunoprecipitated with anti-fibulin-1 antibody, and the immunoprecipitates were analyzed with anti-calumenin-1/2 and anti-fibulin-1 antibodies by Western blot, using horseradish peroxidase (HRP)-conjugated protein A as the secondary antibody. (d) In vitro binding assay of calumenin-1-EGFP (purified with anti-EGFP antibody) and fibulin-1-HA (purified with anti-HA antibody). The purified proteins were mixed and immunoprecipitated with anti-EGFP antibody. The immunoprecipitates were then analyzed by western blot with anti-HA antibody. (e) Conditioned medium from HeLa cells overexpressing calumenin-1-Δ13/14-EGFP was immunoprecipitated with anti-EGFP antibody, and the immunoprecipitates were analyzed with anti-fibulin-1 antibody by western blot. (f) Mapping the interacting domains of calumenin-2-glutathione S-transferase (GST) and EGFP-tagged fibulin-1 truncates. HEK293T cells overexpressing full-length calumenin-2-GST and EGFP-tagged fibulin-1 truncates were immunoprecipitated with anti-EGFP antibody, and analyzed with anti-GST antibody by western blot. Asterisks indicate bands of fibulin-1 truncates. Schematic pictures of fibulin-1 truncates are shown in the left panel. +/−, binding positive/negative. 293T, HEK293T cells; Calu, calumenin; CM, conditioned medium; Co.St., Coomassie blue staining; IP, immunoprecipitation; SP19, signal peptide (amino acids 1–19) of calumenin; TCL, total cell lysates.
… 
Calumenin stabilizes fibulin-1, and both calumenin and fibulin-1 protein levels are inversely correlated with HCC and pancreatic carcinoma. (a) Transwell assays of HeLa cells in the presence of calumenin-1-IRES-EGFP from HEK293T cells, and simultaneously treated with normal IgG or anti-fibulin-1 antibody. Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). Data are mean±s.d., **P<0.01, ***P<0.001, determined by unpaired two-tailed Student’s t-test. (b) HeLa cells were transfected with SP19-EGFP or calumenin-1-EGFP, and simultaneously treated with normal IgG or anti-fibulin-1 antibody (5 μg/ml) for 24 h. Total cell lysates were analyzed with anti-pERK1/2 antibody by western blot. (c) B16/F10 cells stably co-expressing fibulin-1 shRNA and calumenin-1 were injected into C57BL/6J mice via the tail veins. Representative lungs and quantification of the numbers of metastatic pulmonary colonies 2 weeks post-injection are shown (n=15 or 13). Data are mean± s.e.m., n.s., not significant, determined by unpaired two-tailed Student’s t-test. (d, e) HeLa cells were transfected with control or calumenin-1/2 siRNA for 72 h (d), or treated with normal IgG or anti-calumenin-1/2 antibody (5 μg/ml) for 24 h (e). Total cell lysates and conditioned medium were analyzed with anti-calumenin and anti-fibulin-1 antibodies by western blot. (f, g) HeLa cells were transfected with calumenin-1/2-IRES-EGFP (f) or calumenin-1-Δ13/14-EGFP (g) for 48 h. Fibulin-1 in conditioned medium were analyzed by western blot. (h, j) Immunohistochemical staining of calumenin-1/2 and fibulin-1 in HCC tissues (h), pancreatic carcinoma tissues (j) and their corresponding adjacent normal tissues. Scale bars, 100 μm. (i, k) Statistical analysis of the immunohistochemical staining signal intensity of calumenin-1/2 and fibulin-1 in tumor samples from 75 HCC patients (i) and 74 pancreatic tumor patients (k). ↓/↑, expression level decreased/increased in tumor tissue; −, no clear change detected; green box, simultaneous decrease of both calumenin-1/2 and fibulin-1. 293T, HEK293T cells; Calu, calumenin; CM, conditioned medium; Co.St., Coomassie blue staining; SP19, signal peptide (amino acids 1–19) of calumenin; TCL, total cell lysates.
… 
Content may be subject to copyright.
ORIGINAL ARTICLE
Extracellular calumenin suppresses ERK1/2 signaling and cell
migration by protecting bulin-1 from MMP-13-mediated
proteolysis
Q Wang
1,3
, B Shen
1,3
, L Chen
1,3
, P Zheng
1,3
, H Feng
1
, Q Hao
1
, X Liu
1
, L Liu
1
,SXu
1
, J Chen
1,2
and J Teng
1
Extracellular proteins are vital for cell activities, such as cell migration. Calumenin is highly conserved among eukaryotes, but its
functions are largely unclear. Here, we identify extracellular calumenin as a suppressor of cell migration and tumor metastasis.
Calumenin binds to and stabilizes bulin-1, leading to inactivation of extracellular signal-regulated kinases 1 and 2 (ERK1/2)
signaling. We further identify the minimal functional domain of calumenin (amino acids 74138 and 214280). Depletion of
calumenin induces bulin-1- and phospho-ERK1/2 (pERK1/2)-dependent promotion of cell migration. Consistently, in hepatocellular
and pancreatic carcinoma, both calumenin and bulin-1 are downregulated. Furthermore, we show that matrix metalloproteinase-
13 (MMP-13) proteolyzes bulin-1 and that calumenin protects bulin-1 from cleavage by MMP-13. Calumenin, together with
bulin-1, also interacts with bronectin and depends on both syndecan-4 and α5β1-integrin to suppress ERK1/2 signaling and
inhibit cell migration. Thus, extracellular calumenin regulates bulin-1 to have crucial roles in ERK1/2 signaling and cell migration.
Oncogene advance online publication, 17 March 2014; doi:10.1038/onc.2014.52
INTRODUCTION
Cell migration is a fundamental process in embryonic develop-
ment and pathological conditions such as tumor metastasis and
inammation. In the extracellular space, extracellular matrix (ECM)
proteins form a complicated structural and functional network,
which not only provides structural support for cells, but also
integrates complex, multivalent signals to regulate various
physiological processes, like cell migration.
1
Fibulin-1 is a secreted
glycoprotein, which interacts with many other ECM proteins, such
as bronectin, laminin, brinogen, aggrecan and versican,
24
and
is thus considered as an adhesion bridge of ECM structures.
1
Fibulin-1 inhibits cell adhesion and migration via bronectin and
the receptor syndecan-4,
4,5
suppresses angiogenesis possibly
through angiogenin
6,7
and regulates cardiac ventricular morpho-
genesis via versican and ErbB2,
8
suggesting that bulin-1
regulates cell adhesion and migration. However, the molecular
mechanism underlying the functional regulation of bulin-1 in
extracellular space is largely unknown.
Matrix metalloproteinases (MMPs) are the major extracellular
proteinases that regulate ECM homeostasis and remodeling.
9
MMP-13 is a collagenase capable of cleaving rigid collagen brils
to remodel ECM structures, as well as to release embedded
signaling molecules.
10
Besides, MMP-13 also cleaves other
substrates, such as gelatin, aggrecan, perlecan and tenacin-C.
11
The expression of MMP-13 is inuenced by a wide range of
cytokines and hormones, is commonly upregulated in tumors and
is correlated with cancer metastasis and aggressiveness.
1113
Furthermore, MMP-13 is secreted as an immature precursor, and
undergoes activation by auto-proteolysis or peptide removal by
several other MMPs.
14
However, how MMP-13 is regulated
extracellularly after maturing remains to be determined.
Human calumenin, a member of the CREC protein family,
15
is
encoded by the CALU gene (NCBI GeneID: 813).
16
Fifteen
alternative splice variants are identied as calumenin-115, with
calumenin-1/2 (shown as calumenin without specic indication)
being the most abundant.
1719
Calumenin contains a signal
peptide and is detectable in the extracellular space.
20,21
Proteomic
analysis suggests that calumenin is downregulated in MHCC97-H
cells, a human hepatocellular carcinoma (HCC) cell line with high
metastatic potential, compared with low metastatic potential
MHCC97-L cells.
22
Similarly, a lower expression level of calumenin
was also reported in human head and neck squamous cell
carcinoma
23
and lung squamous cell carcinoma.
24
Nevertheless,
the physiological function of extracellular calumenin, and whether
there is a correlation between calumenin expression and tumor
malignancy remain unclear. Here we show that calumenin
protects bulin-1 from MMP-13-mediated proteolysis, and thus
inhibiting extracellular signal-regulated kinases 1 and 2 (ERK1/2)
phosphorylation and cell migration.
RESULTS
Extracellular calumenin inhibits cell migration
We rst tested the effect of calumenin on cell migration.
Overexpression of calumenin-1/2-enhanced green uorescent
protein (EGFP) markedly inhibited wound healing in HeLa
cells (Supplementary Figure S1a). Mutation of asparagine to alanine
at position 131 (N131A) of calumenin-2, which was reported to be
1
State Key Laboratory of Bio-Membrane and Membrane Bio-Engineering, Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, Department of Cell
Biology and Genetics, College of Life Sciences, Peking University, Beijing, China and
2
Center for Quantitative Biology, Peking University, Beijing, China. Correspondence: Dr J Chen
or Dr 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.
E-mail: chenjg@pku.edu.cn or junlinteng@pku.edu.cn
3
These authors contributed equally to this work.
Received 31 October 2013; revised 6 January 2014; accepted 20 January 2014
Oncogene (2014), 113
© 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14
www.nature.com/onc
an N-glycosylation site
20
and important for its stability,
21
resulted in
failure to inhibit wound healing (Supplementary Figure S1a). To test
whether it is the extracellular calumenin that impaired cell
migration, we added conditioned medium from calumenin-1/2-
EGFP-expressing HEK293T cells to HeLa cells, and this also inhibited
wound healing of HeLa cells (Figures 1a and b), while had no
detectable inuence on cell proliferation indicated by BrdU labeling
(Supplementary Figure S1b). Transwell assays conrmed that
extracellular calumenin-1/2-EGFP suppressed cell migration of
HeLa (Figure 1c) and MDA-MB-231 cells (Supplementary Figure
S1c). To exclude the potential steric hindrance of the C-terminal
EGFP tag, we used calumenin-1/2-internal ribosome entry site
(IRES)-EGFP in Matrigel-coated transwell assays, and found that the
numbers of HeLa cells migrated through the transwell were also
signicantly decreased (Figure 1d). Furthermore, addition of
puried calumenin-1-His-Strep to the medium of HeLa cells further
substantiated that extracellular calumenin suppressed cell migra-
tion (Figure 1e).
Calumenin inhibits cell migration by bulin-1
Q Wang et al
2
Oncogene (2014), 1 13 © 2014 Macmillan Publishers Limited
To investigate the effect of calumenin on cell migration and
invasion in vivo, we used the classical B16 melanoma mouse
model.
25
We rst generated B16/F10 melanoma cell lines
stably expressing calumenin-1/2-IRES-EGFP (Supplementary
Figure S1d). Wound-healing assays showed that calumenin-1/2
overexpression also suppressed B16/F10 cell migration
(Supplementary Figure S1e). We then injected these cells into
mice through the tail vein and observed their lungs 2 weeks
later. The number of pulmonary metastatic colonies formed by
calumenin-1/2-overexpressing cells was signicantly fewer than
that by the control cells (Figure 1f and Supplementary Figure S1f),
while no signicant difference in the xenograft tumor
weight (Supplementary Figure S1g) and volume (Supplementary
Figure S1h) were observed when subcutaneously transplanting
these cell lines into nude mice.
Then, loss-of-function of endogenous calumenin-1/2, either
by blockade with its antibody (Figure 1g) or by knockdown with
small interfering RNA (siRNA; Figures 1h and i) in HeLa cells,
markedly promoted cell migration, while addition of condi-
tioned medium containing calumenin-1/2 signicantly rescued
the phenotype induced by knockdown of calumenin-1/2
(Figure 1h and i). Consistently, knockdown of calumenin-1/2 in
B16/F0 cells also promoted cell migration (Supplementary
Figures S2ac), and increased the number of pulmonary
metastatic colonies (Figure 1j). Together, these ndings reveal
that extracellular calumenin suppresses cell migration both
in vitro and in vivo.
Minimal functional domain of calumenin for inhibiting cell
migration
To determine the minimal functional domain of calumenin-1, we
rst tested the effects of 15 isoforms of calumenin on cell
migration, and found that calumenin-1, 2, 3, 4, 5 and 7
inhibited HeLa cell migration (Supplementary Figure S3a). After
analyzing the exon composition of these isoforms, we further
generated 14 EGFP-tagged truncates, named calumenin-1-Δ1to
calumenin-1-Δ14 (Figure 2a), and transfected them into HeLa
cells. Wound-healing assays showed that these truncates fell
into two categories, one inhibiting cell migration like the
full-length calumenin-1-EGFP, while the other having no
signicant effect (Figures 2b and d). The two shortest truncates,
calumenin-1-Δ13 (amino acids 74138 and 214315) and
calumenin-1-Δ14 (amino acids 74138 and 214280) were
conrmed to suppress cell migration by transwell assays
(Figure 2e). To validate the identied minimal functional domain
of calumenin-1 in vivo, we generated B16/F10 melanoma cell
lines stably expressing calumenin-1-Δ14-EGFP (Supplementary
Figure S3b), which showed reduced wound-healing efciency
(Supplementary Figure S3c). We then injected them into mice
through the tail vein. The number of pulmonary metastatic
colonies formed by these cells was signicantly fewer than that
of the control group (Figure 2f and Supplementary Figure S3d).
Thus, the minimal functional domain, calumenin-1-Δ14, is
sufcient to suppress cell migration.
Extracellular calumenin inhibits cell migration via suppressing
ERK1/2 phosphorylation
The mitogen-activated protein kinase signaling pathway is
known as a main signaling pathway to regulate cell
migration,
26
thus we tested the three major mitogen-activated
protein kinases: ERK1/2, p38 and JNK. Overexpression of
calumenin-1/2 suppressed the level of phospho-ERK1/2 (pERK1/2),
but not p-p38 or pJNK, in HeLa (Figure 3a) and B16/F10
melanoma cells (Figure 3b). Consistently, both siRNA knock-
down (Figure 3c) and antibody blockade of calumenin-1/2
(Figure 3d) promoted ERK1/2 phosphorylation, and the
pERK1/2 level in siRNA-treated cells was restored by adding
conditioned medium containing calumenin-1/2 (Figure 3c).
In addition, a decrease of pERK1/2 was detected 7 h
after addition of the eukaryotically expressed and puried
calumenin-1 to the medium of HeLa cells (Figure 3e).
Similar results were observed for pMEK1/2 (Figure 3f), the
upstream kinase of ERK1/2. We then asked whether the
inhibitory effect of calumenin on cell migration was
pERK1/2 dependent. In the presence of U0126, an inhibitor of
ERK1/2 phosphorylation,
27
blockade of calumenin by its
antibody failed to promote cell migration (Figure 3d). Moreover,
the minimal functional domain of calumenin, calumenin-1-Δ13
and calumenin-1-Δ14, also decreased the pERK1/2 level
(Figure 3g). Together, these data suggest that the inhibition of
cell migration by extracellular calumenin is dependent on
ERK1/2 activation.
ERK1/2 regulates actin polymerization and stimulates the
formation of lamellipodia to promote cell migration.
28,29
To
examine the effect of calumenin on cell morphology, we labeled
actin laments along with paxillin, a marker of focal adhesion.
30
Figure 1. Extracellular calumenin suppresses cell migration. (a) Images of wound-healing assays of HeLa cells at 0, 24 and 48 h post-
scratching, in the presence of conditioned medium containing HEK293T-cell-derived SP19-EGFP or calumenin-1/2-EGFP. EGFP-tagged
protein in conditioned medium of HEK293T cells was detected by western blot using anti-EGFP antibody at 48 h post-transfection.
(b)Quantication of wound sizes from (a)(n=8). (c) Transwell assays of HeLa cells, with HEK293T cells in the lower chamber transfected
with SP19-EGFP or calumenin-1/2-EGFP. Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). EGFP-tagged
protein in conditioned medium from HEK293T cells were detected by western blot using anti-EGFP antibody at 48 h post-transfection.
(d) Invasion assays of HeLa cells with HEK293T cells in the lower chamber expressing IRES-EGFP or calumenin-1/2-IRES-EGFP. Numbers of
HeLa cells migrated to the lower side of the transwells are shown (n=4). Calumenin-1/2 in conditioned medium from HEK293T cells were
detected by western blot at 48 h post-transfection. (e) Transwell assays of HeLa cells treated with eukaryotically expressed and puried
calumenin-1-His-Strep (1 μg/ml). Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). (f) B16/F10 cells
stably expressing IRES-EGFP or calumenin-1/2-IRES-EGFP were injected into C57BL/6J mice via the tail veins. Representative lungs and
quantication of the numbers of metastatic pulmonary colonies 2 weeks post-injection are shown (n=12). (g) Transwell assays of HeLa cells
treated with normal IgG or anti-calumenin-1/2 antibody (5 μg/ml). Numbers of HeLa cells migrated to the lower side of the transwells are
shown (n=4). (h) RNA interference efciency of HeLa cells transfected with control or calumenin-1/2 siRNA for 72 h. HEK293T cells derived
calumenin-1/2-containing conditioned medium was concentrated and added for rescue experiments. Calumenin-1/2 in total cell lysates
and conditioned medium were detected by western blot. (i) Transwell assays of HeLa cells corresponding to (h). Numbers of HeLa cells
migrated to the lower side of the transwells are shown (n=4). (j) B16/F0 cells stably expressing control or calumenin-1/2 short hairpin RNA
(shRNA) were injected into C57BL/6J mice via the tail veins. Representative lungs and quantication of the numbers of metastatic
pulmonary colonies 3 weeks post-injection are shown (n=15 or 14). For a,c,d,e,g,i, scale bars, 100 μm; for f,j, scale bars, 1 cm. 293T,
HEK293T cells; Calu, calumenin; CM, conditioned medium; Co.St., Coomassie blue staining; SP19, signal peptide (amino acids 119) of
calumenin; TCL, total cell lysates. For be,g,i,dataaremean±s.d.; for f,j,dataaremean±s.e.m., *Po0.05, **Po0.01, ***Po0.001,
determined by unpaired two-tailed Studentst-test.
Calumenin inhibits cell migration by bulin-1
Q Wang et al
3
© 2014 Macmillan Publishers Limited Oncogene (2014), 1 13
In both HeLa (Figure 3h) and B16/F10 cells (Figures 3h and i),
the elevation of calumenin-1/2 protein level induced a
smooth cell periphery and promoted the cells to aggregate in
a cobblestone-like formation. Conversely, calumenin-1/2 knock-
down in B16/F0 cells resulted in an elongated broblast-like
morphology with pseudopodia-like protrusions (Figures 3j and k).
Thus, calumenin induces cell migration-related morphology
changes.
Calumenin interacts with bulin-1
To screen for calumenin-interacting proteins involved in the
ERK1/2 pathway, in the extracellular space or on the plasma
membrane, we performed immunoprecipitation assays of
total cell lysates and conditioned medium of HEK293T cells
expressing calumenin-1-EGFP. The bound proteins were
separated by sodium dodecyl sulfatepolyacrylamide gel
electrophoresis and analyzed by liquid chromatography with
Figure 2. The minimal functional truncate of calumenin. (a) Schematic pictures of the architecture of 14 calumenin-1 truncates. aa,
amino-acid; +/, truncates suppressing/not suppressing cell migration in wound-healing assays. (b) Expression levels of 14 calumenin-1-
EGFP truncates in HeLa cells were detected by western blot using anti-EGFP antibody or reverse-transcript PCR (RTPCR) 24 h
post-transfection. (c) Wound-healing assays of HeLa cells expressing the EGFP-tagged calumenin-1 truncates at 0, 12, 24, 48 and 72 h
post-scratching. Scale bar, 100 μm. (d)Quantication of wound sizes from (c)(n=8). (e) Transwell assays of HeLa cells, with HEK293T cells
in the lower chamber transfected with calumenin-1-EGFP or calumenin-1-Δ13/14-EGFP. Numbers of HeLa cells migrated to the lower side
of the transwells are shown (n=4). EGFP-tagged protein in conditioned medium from HEK293T cells was detected by
western blot using anti-EGFP antibody at 48 h post-transfection. (f) B16/F10 cells stably expressing SP-EGFP or calumenin-1-Δ14-EGFP
were injected into C57BL/6J mice via the tail veins. Representative lungs and quantication of the numbers of metastatic pulmonary
colonies 2 weeks post-infection are shown (n=13). Scale bar, 1 cm. Calu, calumenin; SP19, signal peptide (amino acids 119)
of calumenin. For d,e, data are mean ±s.d.; for f,dataaremean±s.e.m., *Po0.05, ***Po0.001, determined by unpaired two-tailed
Studentst-test.
Calumenin inhibits cell migration by bulin-1
Q Wang et al
4
Oncogene (2014), 1 13 © 2014 Macmillan Publishers Limited
tandem mass spectrometry. Many potential binding proteins
were identied (Supplementary Figures S4ac), among which
bulin-1 was the most abundant (Figure 4a). As it was also
reported to inhibit ERK1/2 phosphorylation,
4
we focused on
bulin-1.
Fibulin-1 has four isoforms produced by alternative mRNA
splicing (Supplementary Figure S4b) with bulin-1D being the
longest.
31
We then used bulin-1D (described as bulin-1, unless
otherwise indicated) and conrmed its interaction with calum-
enin-1/2 by immunoprecipitation in conditioned medium (Figures
4b and c). Furthermore, both in vitro binding assays (Figure 4d)
and yeast two-hybrid assays (Supplementary Figure S4d) showed
that calumenin-1/2 directly interacted with bulin-1. In addition,
the minimal functional domain of calumenin, calumenin-1-Δ13
and calumenin-1-Δ14, also interacted with bulin-1 (Figure 4e),
suggesting that calumenin-1-Δ14 (amino acids 78134 and
218280) are sufcient for its interaction with bulin-1. However,
the diminished interaction of calumenin minimal functional
truncates and bulin-1 suggested that the other parts of
calumenin may also facilitate its interaction with bulin-1
(Figure 4e). We then generated EGFP-tagged truncation mutants
of bulin-1 to map its interaction domain with calumenin.
Calumenin-2-glutathione S-transferase co-immunoprecipitated
with the full-length bulin-1 and its truncates bulin-1Δ4,
Δ5 and Δ6 (Figure 4f). All these truncates contained EGF-like
repeats 79, suggesting these repeats (amino acids 441578) of
bulin-1 are crucial for its interaction with calumenin. Collectively,
calumenin directly interacts with bulin-1 in the extracellular
space, and amino acids 78134 and 218280 of calumenin and
EGF-like repeats 79ofbulin-1 are required for their interaction.
Calumenin stabilizes bulin-1 and both calumenin and bulin-1
are inversely correlated with hepatocellular and pancreatic
carcinoma
Next, we tested the relationship between calumenin and bulin-1
in cell migration. Blockade of bulin-1 by its antibody signicantly
reduced the suppressive activity of calumenin-1 on HeLa cell
migration (Figure 5a), and effectively blocked the calumenin-1-
induced reduction of ERK1/2 phosphorylation (Figure 5b). More-
over, after knockdown of bulin-1, no signicant difference was
detected between control and calumenin-1-overexpressing B16/F10
cells in wound-healing assays (Supplementary Figures S4e and f).
Consistently, the number of pulmonary metastatic colonies
formed by these cells did not show any signicant difference
(Figure 5c). All these data suggest that calumenin suppresses cell
migration in a bulin-1-dependent manner.
We noticed that the suppressive ability on cell migration was
more efcient in cells co-expressing bulin-1 and calumenin-1
than the expression of each alone (Supplementary Figure S4g),
implying that calumenin and bulin-1 function synergistically to
inhibit cell migration. Interestingly, we found that depletion of
calumenin-1/2 either by siRNA (Figure 5d and Supplementary
Figure S2a) or antibody (Figure 5e) reduced the bulin-1 protein
level, whereas overexpression of calumenin-1/2 markedly
enhanced it in the extracellular space (Figures 3b and 5f), with
its mRNA level unchanged (Supplementary Figure S4h). The
minimal functional domain of calumenin, calumenin-1-Δ13 and
calumenin-1-Δ14, also enhanced the bulin-1 protein level
(Figure 5g). Together, these data suggest that calumenin stabilizes
bulin-1 in the extracellular space.
Proteins that inhibit cell migration are frequently downregu-
lated in cancer.
32
Considering that both calumenin and bulin-1
inhibit cell migration, we sought to determine whether they were
simultaneously downregulated in cancer cells and tumor tissues
from patients. Both the calumenin-1/2 and bulin-1 protein levels
were downregulated, while the pERK1/2 level was elevated in
high metastatic potential MHCC97-H cells, compared with low
metastatic potential MHCC97-L cells (Supplementary Figure S5a).
Consistently, immunohistochemical staining showed decreased
calumenin-1/2 and bulin-1 in most HCC tissues (Figure 5h). In
detail, in samples from 75 HCC patients (Supplementary Figure
S5b and Supplementary Table S1), ~ 65% showed a decrease of
both calumenin-1/2 and bulin-1 in tumor tissues (Figure 5i).
Moreover, patients with higher HCC grades (IIIII) had lower
protein levels of both calumenin-1/2 and bulin-1 than those with
lower grades (III) (Supplementary Figures S5c-e). Similar to HCC,
both calumenin-1/2 and bulin-1 displayed clearly lower expres-
sion in pancreatic tumor tissues compared with their adjacent
normal tissues (Figure 5j and Supplementary Table S2). In samples
from 74 patients with pancreatic carcinoma, ~ 54% showed a
decrease of both proteins (Figure 5k and Supplementary Figure
S5f). Together, these results reveal that both calumenin and
bulin-1 are signicantly downregulated in HCC and pancreatic
carcinoma.
Calumenin protects bulin-1 from cleavage by MMP-13
ECM proteins are usually proteolyzed by MMPs,
9
leading us to
hypothesize that calumenin protects bulin-1 from MMP-
mediated proteolysis. However, no MMP has been identied to
cleave bulin-1. Therefore, we rst overexpressed several MMPs
(MMP-9, -13, -14, -15 and -19) in HeLa cells, and found that only
MMP-13 overexpression decreased the extracellular bulin-1
protein level (Figure 6a). Consistently, in vitro cleavage assays
using the puried proteins also showed that MMP-13ag
cleaved bulin-1-HA, leading to the appearance of specic cleaved
bands (Figure 6b). Therefore, these data suggest that bulin-1 is a
substrate of MMP-13.
To investigate the effect of calumenin on MMP-13-mediated
bulin-1 degradation, we co-overexpressed calumenin-1 and
MMP-13, and found that calumenin-1 suppressed the cleavage
of bulin-1 protein by MMP-13 (Figure 6c). Meanwhile, neither
MMP-13 nor calumenin-1 inuenced the mRNA level of bulin-1
(Supplementary Figure S4h). Further in vitro cleavage assays also
demonstrated that the cleavage of bulin-1 by MMP-13 was
suppressed in the presence of calumenin-1-EGFP (Figure 6d).
To further conrm the relationships of calumenin, bulin-1 and
MMP-13 on cell migration, we performed transwell assays.
MMP-13 enhanced cell migration, while blockade of bulin-1 by
its antibody eliminated this enhancement, suggesting that
MMP-13 functions in a bulin-1-dependent manner (Figure 6e).
Importantly, overexpression of calumenin-1-EGFP abolished the
MMP-13-mediated promotion of cell migration (Figure 6f).
Together, these data reveal that MMP-13 degrades bulin-1 to
facilitate cell migration, while calumenin binds to bulin-1 and
protects it from MMP-13-induced cleavage to inhibit cell
migration.
Calumenin inhibits cell migration through bronectin, syndecan-4
and α5β1-integrin
Fibulin-1 was reported to inhibit cell migration via its association
with bronectin,
4
leading us to investigate the relationships
among bronectin, bulin-1 and calumenin. Overexpressed
calumenin-1-EGFP interacted with both endogenous bulin-1
and bronectin (Figure 4b), and endogenous calumenin-1/2 and
bulin-1 were co-immunoprecipitated with bronectin (Figure 7a),
suggesting that calumenin forms a complex with bulin-1 and
bronectin. Besides, calumenin-1/2-EGFP overexpression led to
more bulin-1 co-immunoprecipitated with bronectin
(Figure 7b). Blockade of calumenin-1/2 by its antibody facilitated
cell migration, however, in the presence of anti-bronectin
antibody, anti-calumenin-1/2 antibody failed to enhance cell
migration (Figure 7c), suggesting that bronectin is indispensable
for calumenin and bulin-1 to inhibit cell migration.
Calumenin inhibits cell migration by bulin-1
Q Wang et al
5
© 2014 Macmillan Publishers Limited Oncogene (2014), 1 13
Previous work showed that bulin-1 inhibits broblast cell
adhesion through competing with syndecan-4 to bind to
bronectin, thus inhibiting cell adhesion and migration.
5
There-
fore, we examined whether syndecan-4 is required for the
function of calumenin to inhibit cell migration. The enhanced cell
migration induced by anti-calumenin-1/2 antibody treatment was
abolished by adding anti-syndecan-4 antibody to the medium
(Figure 7d). Moreover, the effect of antibody against calumenin on
Calumenin inhibits cell migration by bulin-1
Q Wang et al
6
Oncogene (2014), 1 13 © 2014 Macmillan Publishers Limited
cell migration was also antagonized by antibodies against α5β1-
integrin (Figure 7e), a major cell surface receptor of bronectin
33
that synergistically function with syndecans in cell migration and
adhesion.
34,35
Although more studies are needed to address their
relationships, these data suggest that the inhibition of cell
migration by calumenin depends on bronectin, syndecan-4 and
α5β1-integrin.
In conclusion, calumenin directly interacts with bulin-1,
protects it from cleavage by MMP-13, and enhances the binding
of bulin-1 to bronectin. Through syndecan-4 and α5β1-integrin
receptors, the formation of a calumenin/bulin-1/bronectin
complex leads to inhibition of the ERK1/2 signaling pathway
and the subsequent suppression of cell migration (Figure 7f).
DISCUSSION
ECM remodeling is a key event of cell migration, and the activity of
extracellular proteases, especially MMPs, has crucial roles in this
process.
9
Among MMPs, MMP-13 is highly expressed in tumor and
stroma tissues, and is a suitable target of anti-cancer and anti-
inammation drugs.
13,36,37
Previous studies showed that MMP-13
activity is mainly regulated at three distinct levels: transcription,
Figure 3. Calumenin suppresses ERK1/2 phosphorylation and regulates actin organization. (a,b) Western blot of total cell lysates and
conditioned medium from HeLa (a) or B16/F10 (b) cells overexpressing the indicated proteins for 48 h. Quantication of relative pERK1/2 to
ERK1/2 levels (a) are shown (n=3). (c) Western blot of total cell lysates and conditioned medium from HeLa cells at 72 h post-transfection with
control or calumenin-1/2 siRNA. HEK293T cells derived calumenin-1/2-containing conditioned medium was concentrated and added for
rescue experiments. (d) Transwell assays of HeLa cells treated with MEK inhibitor U0126 (10 μM) and anti-calumenin-1/2 antibody (5 μg/ml).
Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). Protein levels of total cell lysates from the treated HeLa
cells were analyzed by western blot using anti-pERK1/2 and anti-calumenin antibodies. (e,f) pERK1/2 (e) and pMEK1/2 (f) of HeLa cells treated
with eukaryotically expressed and puried calumenin-1-His-Strep (1 μg/ml) were detected by western blot. (g) pERK1/2 of HeLa cells
overexpressing calumenin-1-EGFP or calumenin-1-Δ13/14-EGFP at 24 h post-transfection were detected by western blot. (h) Co-staining of
paxillin and actin laments in HeLa cells overexpressing SP19-EGFP or calumenin-1/2-EGFP for 72 h. (i,j) Co-staining of paxillin and actin
laments (i) or differential interference contrast images (j) of B10/F10 cells stably expressing IRES-EGFP or calumenin-1-IRES-EGFP.
(k,l) Co-staining of paxillin and actin laments (k) or differential interference contrast images (l) of B10/F0 cells stably expressing control short
hairpin RNA (shRNA) or calumenin-1/2 shRNA. For h,i,k, scale bars, 10 μm; for j,l, scale bars, 100 μm. Calu, calumenin; CM, conditioned
medium; Co.St., Coomassie blue staining; SP19, signal peptide (amino acids 119) of calumenin-1/2; TCL, total cell lysates. For a,d, data are
mean ±s.d., **Po0.01, ***Po0.001, determined by unpaired two-tailed Studentst-test.
Figure 4. Calumenin interacts with bulin-1 in the extracellular space. (a) Immunoprecipitation assays with anti-EGFP antibody in conditioned
medium and total cell lysates from HEK293T cells expressing calumenin-1-EGFP. Immunoprecipitates were subjected to sodium dodecyl
sulfatepolyacrylamide gel electrophoresis (SDSPAGE) and Coomassie blue staining. Arrows indicate the proteins identied by liquid
chromatography with tandem mass spectrometry (LC-MS/MS) analysis. (b) Conditioned medium from HEK293T cells expressing SP19-EGFP or
calumenin-1/2-EGFP was immunoprecipitated with anti-EGFP antibody, and the immunoprecipitates were analyzed with anti-bulin-1, anti-
bronectin and anti-EGFP antibodies by western blot. (c) Conditioned medium from HeLa cells was immunoprecipitated with anti-bulin-1
antibody, and the immunoprecipitates were analyzed with anti-calumenin-1/2 and anti-bulin-1 antibodies by Western blot, using horseradish
peroxidase (HRP)-conjugated protein A as the secondary antibody. (d)In vitro binding assay of calumenin-1-EGFP (puried with anti-EGFP
antibody) and bulin-1-HA (puried with anti-HA antibody). The puried proteins were mixed and immunoprecipitated with anti-EGFP
antibody. The immunoprecipitates were then analyzed by western blot with anti-HA antibody. (e) Conditioned medium from HeLa cells
overexpressing calumenin-1-Δ13/14-EGFP was immunoprecipitated with anti-EGFP antibody, and the immunoprecipitates were analyzed with
anti-bulin-1 antibody by western blot. (f) Mapping the interacting domains of calumenin-2-glutathione S-transferase (GST ) and EGFP-tagged
bulin-1 truncates. HEK293T cells overexpressing full-length calumenin-2-GST and EGFP-tagged bulin-1 truncates were immunoprecipitated
with anti-EGFP antibody, and analyzed with anti-GST antibody by western blot. Asterisks indicate bands of bulin-1 truncates. Schematic
pictures of bulin-1 truncates are shown in the left panel. +/ , binding positive/negative. 293T, HEK293T cells; Calu, calumenin; CM,
conditioned medium; Co.St., Coomassie blue staining; IP, immunoprecipitation; SP19, signal peptide (amino acids 119) of calumenin; TCL,
total cell lysates.
Calumenin inhibits cell migration by bulin-1
Q Wang et al
7
© 2014 Macmillan Publishers Limited Oncogene (2014), 1 13
post-translational activation and interaction with inhibitors.
11,38
However, as far as we know, no protein has been reported to bind
to MMP-13 substrates to inhibit MMP-13-mediated cleavage. Here,
we show that bulin-1 cleavage by MMP-13 is blocked by
calumenin. Whether calumenin directly inhibits MMP-13 activity,
or protects bulin-1 from MMP-13 access remains to be
investigated. Nevertheless, our ndings open a new dimension
in the understanding of the regulation of MMP-13. We propose
that combined treatment with calumenin and inhibitors of
MMP-13 may serve as a new therapeutic strategy for tumor
metastasis. Besides, calumenin interacts with bronectin, which is
also a substrate of MMP-13.
39
It is possible that calumenin also
inhibits MMP-13-mediated proteolysis of bronectin to inhibit cell
migration, which needs further investigation.
Fibulin-1 assists in the formation of macromolecular ECM
structures,
1
and also inhibits cell migration.
4
Here, we rst
identify bulin-1 as a substrate of MMP-13, and detect MMP-13-
digested bands of bulin-1 (Figure 6b). We propose that multiple
Figure 5. Calumenin stabilizes bulin-1, and both calumenin and bulin-1 protein levels are inversely correlated with HCC and pancreatic
carcinoma. (a) Transwell assays of HeLa cells in the presence of calumenin-1-IRES-EGFP from HEK293T cells, and simultaneously treated with
normal IgG or anti-bulin-1 antibody. Numbers of HeLa cells migrated to the lower side of the transwells are shown (n=4). Data are mean ±s.d.,
**Po0.01, ***Po0.001, determined by unpaired two-tailed Studentst-test. (b) HeLa cells were transfected with SP19-EGFP or calumenin-1-
EGFP, and simultaneously treated with normal IgG or anti-bulin-1 antibody (5 μg/ml) for 24 h. Total cell lysates were analyzed with anti-
pERK1/2 antibody by western blot. (c) B16/F10 cells stably co-expressing bulin-1 shRNA and calumenin-1 were injected into C57BL/6J mice
via the tail veins. Representative lungs and quantication of the numbers of metastatic pulmonary colonies 2 weeks post-injection are shown
(n=15 or 13). Data are mean±s.e.m., n.s., not signicant, determined by unpaired two-tailed Studentst-test. (d,e) HeLa cells were transfected
with control or calumenin-1/2 siRNA for 72 h (d), or treated with normal IgG or anti-calumenin-1/2 antibody (5 μg/ml) for 24 h (e). Total cell
lysates and conditioned medium were analyzed with anti-calumenin and anti-bulin-1 antibodies by western blot. (f,g) HeLa cells were
transfected with calumenin-1/2-IRES-EGFP (f) or calumenin-1-Δ13/14-EGFP (g) for 48 h. Fibulin-1 in conditioned medium were analyzed by
western blot. (h,j) Immunohistochemical staining of calumenin-1/2 and bulin-1 in HCC tissues (h), pancreatic carcinoma tissues (j) and their
corresponding adjacent normal tissues. Scale bars, 100 μm. (i,k) Statistical analysis of the immunohistochemical staining signal intensity of
calumenin-1/2 and bulin-1 in tumor samples from 75 HCC patients (i) and 74 pancreatic tumor patients (k). /, expression level decreased/
increased in tumor tissue; , no clear change detected; green box, simultaneous decrease of both calumenin-1/2 and bulin-1. 293T, HEK293T
cells; Calu, calumenin; CM, conditioned medium; Co.St., Coomassie blue staining; SP19, signal peptide (amino acids 119) of calumenin; TCL,
total cell lysates.
Calumenin inhibits cell migration by bulin-1
Q Wang et al
8
Oncogene (2014), 1 13 © 2014 Macmillan Publishers Limited
MMP-13 cleavage sites exist on bulin-1, which is consistent
with the previous reports that MMPs tend to cleave substrates
at many sites.
40
As these bands (mostly >50 kDa) are recognized
by anti-HA antibody, and the HA epitope is tagged at the
C-terminus of bulin-1, we speculate that some of the MMP-13
cleavage sites are located in its N-terminus. Considering that
calumenin is associated with EGF-like repeats 79 on the
C-terminus of bulin-1, it is unlikely that calumenin simply shields
the MMP-13 cleavage sites on bulin-1, but rather stabilizes its
whole structure.
Figure 6. Calumenin inhibits MMP-13-mediated bulin-1 proteolysis. (a) Fibulin-1 were detected by western blot in conditioned medium of
HeLa cells overexpressing different MMPs. Expression levels of MMPs were detected by RTPCR. (b)In vitro cleavage assays of bulin-1-HA
(puried by anti-HA antibody) by MMP-13ag (puried by anti-ag antibody, and activated by its activator APMA). 1 × , 2× , 4 × , 8× and
16 × indicate increasing concentration of MMP-13. Cleaved bands of bulin-1 are marked by asterisks. (c) HeLa cells were co-expressed with
calumenin-1-EGFP and MMP-13ag for 48 h. Total cell lysates and conditioned medium were analyzed with anti-bulin-1, anti-EGFP, and
anti-ag antibodies by western blot. (d)In vitro cleavage assays of bulin-1-HA (puried by anti-HA antibody) by MMP-13ag (puried by
anti-ag antibody, and activated by its activator APMA), in the presence or absence of calumenin-1-EGFP (puried by anti-EGFP antibody). The
cleavage samples were analyzed with anti-HA antibody by western blot. (e) Transwell assays of HeLa cells overexpressing MMP-13ag,
and simultaneously treated with normal IgG or anti-bulin-1 antibody (5 μg/ml). Numbers of HeLa cells migrated to the lower side of
transwells are shown (n=4). MMP-13ag in conditioned medium from HEK293T cells were detected by western blot using anti-ag
antibody. (f) Transwell assays of HeLa cells co-expressing calumenin-1-EGFP and MMP-13ag. Numbers of HeLa cells migrated to the
lower side of transwells are shown (n=4). EGFP- or ag-tagged protein in conditioned medium from HeLa cells were detected by western
blot using anti-EGFP or anti-ag antibody, respectively. APMA, 4-aminophenylmercuric acetate; Calu, calumenin; CM, conditioned medium;
Co.St., Coomassie blue staining; SP19, signal peptide (amino acids 119) of calumenin; TCL, total cell lysates. For e,f, data are mean ±s.d.,
**Po0.01, ***Po0.001, n.s. not signicant, determined by unpaired two-tailed Studentst-test.
Calumenin inhibits cell migration by bulin-1
Q Wang et al
9
© 2014 Macmillan Publishers Limited Oncogene (2014), 1 13
Our results, as well as previous reports, show that the full-length
bulin-1 inhibits cell migration,
5
while no such inhibition occurs
after its degradation by MMP-13. However, the MMP-digested
fragments of some ECM proteins bind to and activate cell surface
receptors.
33
Therefore, whether the MMP-13-digested fragments
of bulin-1 have regulatory roles in cell migration or in other
physiological process is worth further investigation.
In HCC and pancreatic carcinoma tissues, we nd that
calumenin and bulin-1 are inversely correlated with tumor grade
and metastatic potential. A similar situation of calumenin is also
described in human head and neck squamous cell carcinoma
23
and lung squamous cell carcinoma.
24
A recent report showed that
bulin-1 mRNA and protein levels are reduced in human HCC
samples.
41
These data further strengthen that calumenin and
bulin-1 suppress tumor metastasis. However, progressive expres-
sion of calumenin is reported in colon
42
and endometrial
carcinomas.
43
Similarly, elevated bulin-1 are also detected in
ovarian and breast carcinomas,
44,45
and bulin-1C may participate
in ovarian tumorigenesis,
46
suggesting that the inverse association
between calumenin, bulin-1 and tumor metastasis may be
tissue specic.
MATERIALS AND METHODS
Cell lines and tumor samples
HeLa, HEK293T, MCF-7, MDA-MB-231, MHCC97-L and MHCC97-H cells were
cultured in Dulbeccos modied Eagles medium (GIBCO, Grand Island, NY,
USA), B16/F10 and B16/F0 in RPMI 1640 (GIBCO) and supplemented with
10% fetal bovine serum (GIBCO) at 37 °C with 5% CO
2
.
Reagents and antibodies
Reagents used were: 4-aminophenylmercuric acetate (Merck, Whitehouse
Station, NJ, USA), BrdU (Calbiochem, Darmstadt, Germany), ERK1/2
inhibitor U0126 (Promega, Madison, WI, USA), G418 (GIBCO), horseradish
Figure 7. Calumenin inhibits cell migration through bronectin, syndecan-4 and α5β1-integrin. (a) Conditioned medium from HeLa cells was
immunoprecipitated with anti-bronectin antibody, and the immunoprecipitates were analyzed with anti-bulin-1 and anti-calumenin
antibodies by western blot. (b) Conditioned medium from HEK293T cells overexpressing SP19-EGFP or calumenin-1/2-EGFP was
immunoprecipitated with anti-bronectin antibody. The immunoprecipitates were analyzed by western blot with anti-bulin-1 antibody.
(ce) Transwell assays of HeLa cells in the presence of anti-calu-1/2 antibody (5 μg/ml), together with anti-bronectin antibody (5 μg/ml) (c), or
anti-syndecan-4 antibody (5 μg/ml) (d), or anti-β1 and anti-α5-integrin antibodies (5 μg/ml) (e). Numbers of HeLa cells migrated to the lower
side of transwells are shown (n=4). (f) A schematic model illustrating that calumenin suppresses MMP-13-mediated proteolysis of bulin-1 in
the extracellular space to regulate cell migration. Calumenin-1/2 protects bulin-1 from MMP-13-mediated proteolysis, and the stabilized
bulin-1 binds to bronectin, which possibly decreases the binding of integrins and syndecans-4 to bronectin, and thus inhibits the
syndecan-integrin-ERK1/2 signaling pathway and cell migration. Calu, calumenin; CM, conditioned medium; Co.St., Coomassie blue staining;
IP, immunoprecipitation; SP19, signal peptide (amino acids 119) of calumenin; TCL, total cell lysates. For ce, data are mean ±s.d.,
***Po0.001, n.s. not signicant, determined by unpaired two-tailed Studentst-test.
Calumenin inhibits cell migration by bulin-1
Q Wang et al
10
Oncogene (2014), 1 13 © 2014 Macmillan Publishers Limited
peroxidase-conjugated protein A (Sigma-Aldrich, St Louis, MO, USA), JetPEI
(Polyplus, Illkirch, France), Phalloidin (Atto 647N; Sigma-Aldrich), puromycin
(Sigma-Aldrich), Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) and
Matrigel (BD Biosciences, Franklin Lakes, NJ, USA). Rabbit anti-EGFP
polyclonal antibody were self-produced. Antibodies of α5-integrin (Abcam,
Cambridge, UK, ab75472), β1-integrin (Abcam, ab24693), calumenin
(ProteinTech, Wuhan, China, 178041-AP), BrdU (MBL, MI-113, Woburn,
MA, USA), EGFP (MBL, M0483), ERK1/2 (Santa Cruz, Dallas, TX, USA, sc-154),
phospho-ERK1/2 (Santa Cruz, sc-7383), bronectin (Abcam, ab23750),
bronectin (BD Biosciences, 610078), bulin-1 (Abcam, ab54652), bulin-1
(3A11, was provided by W Scott Argraves),
4
bulin-1 (Santa Cruz, sc-20818),
ag (Sigma-Aldrich, F1804), HA (Sigma-Aldrich, H9658), JNK (Cell Signaling,
Danvers, MA, USA, 9252), phospho-JNK (Cell Signaling, 9251), MEK1/2
(Epitomics, Burlingame, CA, USA, S2436), phospho-MEK1/2 (Epitomics,
14701), p38 (Cell Signaling, 9212), phospho-p38 (Cell Signaling, 9216),
paxillin (BD Biosciences, 610052) and sydecan-4 (Santa Cruz, sc-12766)
were used. The secondary antibodies Alexa Fluor 488-/568-conjugated
goat anti-mouse/rabbit IgG (Invitrogen) and horseradish peroxidase-
conjugated antibodies (Jackson ImmunoResearch, West Grove, PA, USA)
were used.
Vector construction
Human calumenin-1/2 and bulin-1D transcripts were amplied from HeLa
cell complementary DNA. MMP-9, -13, -14, -15 and -19 were amplied from
HeLa or HEK293T cell complementary DNA. Short hairpin RNAs were
constructed into pLKO.1 vector (Sigma-Aldrich).
Wound-healing and two-chamber transwell assays
Wound-healing assays were performed as previously reported.
47
For
transwell assays, 6.5-mm Millicell inserts (Millipore, Billerica, MA, USA) were
used as previously reported.
48
HEK293T or MCF-7 cells in the lower
chamber were transfected with plasmids, and 24 h post-transfection, HeLa
or MDA-MB-231 cells (2 × 10
5
cells) were added into the upper chamber of
each Millicell. For antibody blockade, antibodies (5 μg/ml) were added into
the lower chamber. After 68 h, cells that migrated to the lower surface
were stained with haematoxylin and eosin. Samples were examined under
an IX71 inverted microscope (Olympus, Tokyo, Japan) and statistically
calculated.
RNA interference and lentivirus package
For calumenin knockdown, HeLa cells were transfected with an siRNA pool
against calumenin (see Supplementary Table S3 for target sequences).
Total cell lysates and conditioned medium were collected and subjected to
western blot 72 h after transfection. For rescue assays, calumenin
expressed by HEK293T cells was added into conditioned medium 48 h
post-transfection. For lentivirus production, the pLKO.1 vector containing
short hairpin RNA was transfected into HEK293T cells together with
packaging plasmids, and virus was collected 72 h post-transfection and
used to infect target cells as previously reported.
49
Stable cell line generation
G418 (1 mg/ml) or puromycin (2 μg/ml) was added to B16/F10 or B16/F0
cells transfected with plasmids to kill the negative cells. Single cell with
green uorescence was sorted into 96-well plates by ow cytometry
(Beckman Coulter, Brea, CA, USA) and cultured in the presence of G418 or
puromycin for 12 weeks. The proliferated cells were then subjected to
quantitative real-timePCR and western blot analysis. At least two clones
were established for each cell line.
Animal studies
All animal experiments were undertaken in accordance with the National
Institute of Health Guide for the Care and Use of Laboratory Animals, and
with the approval of the Committee of Animal Research in Peking
University. Animals were kept in a specic pathogen-free environment in
the Peking University Laboratory Animal Center. For pulmonary coloniza-
tion mouse model, experiments were performed as previously described.
50
B16/F10 or B16/F0 stable cell lines (25×10
5
) were slowly injected into the
tail veins of female C57BL/6J mice (7 weeks old). After 2 or 3 weeks, the
in vivo imaging system FX Pro (Kodak, Rochester, NY, USA) was used to
image the EGFP uorescence in living mice whose abdominal hair had
been shaved. The uorescence intensity of the lung region was calculated.
Numbers of melanoma colonies in the lung were counted. For primary
tumor growth mouse model, B16/F10 cells (5 × 10
6
) suspended in Matrigel
were inoculated subcutaneously in the dorsal skinfold of BALB/c nude
mice (6 weeks old). Tumor length and width were measured on days 3, 6,
8, 9, 10, 11, 12 and 13 using calipers, and tumor volume was calculated as
π/6 × length × width
2
. On day 13 after inoculation, the mice were killed and
tumors were surgically removed and weighed.
Protein purication and in vitro experiments
For purication of calumenin-16 × His-Strep, HEK293T cells were trans-
fected with corresponding plasmids and maintained for 24 h followed by
replacement with serum-free Dulbeccos modied Eagles medium for 48 h.
Total cell lysates and conditioned medium were harvested and puried by
sequential binding with Ni-NTA afnity chromatography (Invitrogen) and
Strep-Tactin columns (IBA, Göttingen, Germany).
For purication of calumenin-EGFP, bulin-1-HA and proMMP-13
ag, HEK293T cells transfected with corresponding plasmids were
maintained for 24 h followed by replacement with serum-free Dulbeccos
modied Eagles medium for 48 h. Conditioned medium was harvested
and concentrated using Centricon YM-30 Concentrators (Millipore), and
used for immunoprecipitation. After washing with high-salt buffer (25 mM
HEPES, 2 mMCaCl
2
,1mMMgCl
2
, 500 mMNaCl, pH 7.4), the immunopre-
cipitated protein was eluted with HA or ag peptide, and used for in vitro
assays.
proMMP-13ag was activated by treatment with 1 mM4-
aminophenylmercuric acetate for 45 min at 37 °C. The in vitro cleavage
was carried out in cleavage buffer (50 mMTrisHCl, 10 mMCaCl
2
, 150 mM
NaCl, 0.05% Brij-35, pH 7.5) at 37 °C for 24 h.
Immunouorescence and histochemistry
Cells were xed and labeled as previously described.
51
Paxillin was
immunostained with Alexa Fluor 568-conjugated secondary antibody, and
actin laments were stained with Atto-647N-labeled phalloidin. Samples
were observed under an IX71 inverted uorescence microscope (Olympus)
and a TCS SP2 confocal microscope (Leica, Solms, Germany) equipped with
a × 100/1.4 numerical aperture oil immersion objective lens.
Immunohistochemical staining of samples from HCC and pancreatic
carcinoma patients were performed by the National Engineering Center for
Biochip (Shanghai, China).
Immunoprecipitation, liquid chromatography with tandem mass
spectrometry and western blot
For identifying proteins interacting with calumenin, total cell lysates and
conditioned medium of HeLa cells transfected with calumenin-1-EGFP
were collected 48 h post-transfection. The samples were incubated with
anti-EGFP antibody overnight followed by conjugation with protein A
sepharose (Amersham, Pittsburgh, PA, USA) in immunoprecipitation buffer
(20 mMHEPES, 120 mMNaCl, 2 mMCaCl
2
,1mMMgCl
2
,1mMdithiothreitol,
0.1% Triton X-100, pH 7.4) with proteinase inhibitors. Samples were then
separated by sodium dodecyl sulfatepolyacrylamide gel electrophoresis.
Specic bands were excised and digested with sequence-grade modied
trypsin (Promega) and subjected to LCQ Deca XP Plus Analyzer liquid
chromatography/mass spectrometry (Finnigan, San Diego, CA, USA)
analysis.
Immunoprecipitation assays were performed as previously described.
52
For western blot assays, the proteins were transferred onto polyvinylidene
diuoride membranes (Millipore). Membranes were blocked and then
probed with primary and secondary antibodies.
RNA isolation and quantitative real-timePCR
Total RNA was extracted with Tranzol reagent (Transgen, Beijing, China)
and reverse transcribed (Invitrogen) as in previous reports.
53,54
Quantita-
tive real-timePCR was performed by an ABI 7300 detection system
(Applied Biosystems, Foster City, CA, USA) using SYBR Green PCR Master
Mix (Invitrogen) as previously described.
53
Glyceraldehyde 3-phosphate
dehydrogenase served as a reference control and the 2
ΔΔCT
method
55
was used (see Supplementary Table S3 for primers).
BrdU incorporation assay
Cells were incubated in 15 μMBrdU (Calbiochem) for 1 h before xation,
permeabilized in 1% Triton X-100 and then treated with 1 Mand 2 MHCl
Calumenin inhibits cell migration by bulin-1
Q Wang et al
11
© 2014 Macmillan Publishers Limited Oncogene (2014), 1 13
sequentially. Cells were then neutralized and incubated with anti-BrdU
antibody. Finally, cells were labeled with 4,6-diamidino-2-phenylindole and
visualized under an IX71 inverted uorescence microscope (Olympus).
Yeast two-hybrid assay
Yeast two-hybrid assays were performed using Matchmaker GAL4 two-
hybrid system 3 (Clontech, Mountain View, CA ,USA). pGBKT7-calumenin-2
and pGADT7-bulin-1 were co-transformed into yeast strain AH109. The
yeasts were plated on SC-Trp-Leu plates and positive colonies were
subsequently plated on SC-Trp-Leu-His-Ade plates.
Statistical analysis
Studentst-test was used to analyze data.
CONFLICT OF INTEREST
The authors declare no conict of interest.
ACKNOWLEDGEMENTS
We thank Dr W Scott Argraves at the Medical University of South Carolina for kindly
providing anti-bulin-1 antibody (3A11). The lentivirus system is a kind gift from
Dr Jincai Luo at Peking University. We also thank Dr Li Yu and Dr Xiaofeng Wang at
Tsinghua University, and Dr Zhengfan Jiang at Peking University for helpful
discussion. We also thank Dr IC Bruce at Zhejiang University and Dr Xiaolei Su at
the University of California, San Francisco, 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).
REFERENCES
1 Hynes RO. The extracellular matrix: not just pretty brils. Science 2009; 326:
12161219.
2 Olin AI, Morgelin M, Sasaki T, Timpl R, Heinegard D, Aspberg A. The proteoglycans
aggrecan and Versican form networks with bulin-2 through their lectin domain
binding. J Biol Chem 2001; 276: 12531261.
3 Roark EF, Keene DR, Haudenschild CC, Godyna S, Little CD, Argraves WS.
The association of human bulin-1 with elastic bers: an immunohistological,
ultrastructural, and RNA study. J Histochem Cytochem 1995; 43:401411.
4 Twal WO, Czirok A, Hegedus B, Knaak C, Chintalapudi MR, Okagawa H et al.
Fibulin-1 suppression of bronectin-regulated cell adhesion and motility. J Cell Sci
2001; 114: 45874598.
5 Williams SA, Schwarzbauer JE. A shared mechanism of adhesion modulation for
tenascin-C and bulin-1. Mol Biol Cell 2009; 20:11411149.
6 Xie L, Palmsten K, MacDonald B, Kieran MW, Potenta S, Vong S et al. Basement
membrane derived bulin-1 and bulin-5 function as angiogenesis inhibitors and
suppress tumor growth. Exp Biol Med 2008; 233:155162.
7 Zhang H, Gao X, Weng C, Xu Z. Interaction between angiogenin and bulin 1:
evidence and implication. Acta Biochimica et Biophysica Sinica 2008; 40:375380.
8 Cooley MA, Fresco VM, Dorlon ME, Twal WO, Lee NV, Barth JL et al. Fibulin-1 is
required during cardiac ventricular morphogenesis for versican cleavage,
suppression of ErbB2 and Erk1/2 activation, and to attenuate trabecular cardio-
myocyte proliferation. Dev Dynamics 2012; 241:303314.
9 Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the
tumor microenvironment. Cell 2010; 141:5267.
10 Vandenbroucke RE, Dejonckheere E, Van Hauwermeiren F, Lodens S, De Rycke R,
Van Wonterghem E et al. Matrix metalloproteinase 13 modulates intestinal
epithelial barrier integrity in inammatory diseases by activating TNF. EMBO Mol
Med 2013; 5: 10001016.
11 Leeman MF, Curran S, Murray GI. The structure, regulation, and function of human
matrix metalloproteinase-13. Crit Rev Biochem Mol Biol 2002; 37:149166.
12 Klein T, Bischoff R. Physiology and pathophysiology of matrix metalloproteases.
Amino Acids 2011; 41:271290.
13 Zigrino P, Kuhn I, Bauerle T, Zamek J, Fox JW, Neumann S et al. Stromal expression
of MMP-13 is required for melanoma invasion and metastasis. J Invest Dermatol
2009; 129: 26862693.
14 Knauper V, Will H, Lopez-Otin C, Smith B, Atkinson SJ, Stanton H et al. Cellular
mechanisms for human procollagenase-3 (MMP-13) activation. Evidence that
MT1-MMP (MMP-14) and gelatinase a (MMP-2) are able to generate
active enzyme. J Biol Chem 1996; 271: 1712417131.
15 Honore B. The rapidly expanding CREC protein family: members, localization,
function, and role in disease. Bioessays 2009; 31:262277.
16 Yabe D, Taniwaki M, Nakamura T, Kanazawa N, Tashiro K, Honjo T. Human
calumenin gene (CALU): cDNA isolation and chromosomal mapping to 7q32.
Genomics 1998; 49: 331333.
17 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
+-binding activity. Biochim Biophys Acta 1998; 1386:121131.
18 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;
445:440444.
19 Feng H, Chen L, Wang Q, Shen B, Liu L, Zheng P et al. Calumenin-15 facilitates
lopodia formation by promoting TGF-beta superfamily cytokine GDF-15 tran-
scription. Cell Death Dis 2013; 4: e870.
20 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; 248:473481.
21 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.
22 Ding SJ, Li Y, Shao XX, Zhou H, Zeng R, Tang ZY et al. Proteome analysis of
hepatocellular carcinoma cell strains, MHCC97-H and MHCC97-L, with different
metastasis potentials. Proteomics 2004; 4:982994.
23 Wu W, Tang X, Hu W, Lotan R, Hong WK, Mao L. Identication and validation of
metastasis-associated proteins in head and neck cancer cell lines by two-dimensional
electrophoresis and mass spectrometry. Clin Exp Metastasis 2002; 19:319326.
24 Shen C, Hui Z, Wang D, Jiang G, Wang J, Zhang G. Molecular cloning, identi-
cation and analysis of lung squamous cell carcinoma-related genes. Lung Cancer
2002; 38: 235241.
25 Kapoor A, Goldberg MS, Cumberland LK, Ratnakumar K, Segura MF, Emanuel PO
et al. The histone variant macroH2A suppresses melanoma progression through
regulation of CDK8. Nature 2010; 468: 11051109.
26 Huang C, Jacobson K, Schaller MD. MAP kinases and cell migration. J Cell Sci 2004;
117:46194628.
27 Duncia JV, Santella 3rd JB, Higley CA, Pitts WJ, Wityak J, Frietze WE et al. MEK
inhibitors: the chemistry and biological activity of U0126, its analogs, and cycli-
zation products. Bioorganic Medicinal Chem Lett 1998; 8: 28392844.
28 Mendoza MC, Er EE, Zhang W, Ballif BA, Elliott HL, Danuser G et al. ERK-MAPK
drives lamellipodia protrusion by activating the WAVE2 regulatory complex.
Mol Cell 2011; 41: 661671.
29 Martinez-Quiles N, Ho HY, Kirschner MW, Ramesh N, Geha RS. Erk/Src
phosphorylation of cortactin acts as a switch on-switch off mechanism that
controls its ability to activate N-WASP. Mol Cell Biol 2004; 24: 52695280.
30 Cukierman E, Pankov R, Stevens DR, Yamada KM. Taking cell-matrix adhesions to
the third dimension. Science 2001; 294: 17081712.
31 Pan TC, Kluge M, Zhang RZ, Mayer U, Timpl R, Chu ML. Sequence of extracellular
mouse protein BM-90/bulin and its calcium-dependent binding to other
basement-membrane ligands. Eur J Biochem 1993; 215:733740.
32 Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;
144:646674.
33 Labat-Robert J. Cell-Matrix interactions, the role of bronectin and integrins.
A survey. Pathologie-Biologie 2012; 60:1519.
34 Morgan MR, Humphries MJ, Bass MD. Synergistic control of cell adhesion by
integrins and syndecans. Nat Rev Mol Cell Biol 2007; 8:957969.
35 Morgan MR, Hamidi H, Bass MD, Warwood S, Ballestrem C, Humphries MJ.
Syndecan-4 phosphorylation is a control point for integrin recycling. Dev Cell
2013; 24: 472485.
36 Vizoso FJ, Gonzalez LO, Corte MD, Corte MG, Bongera M, Martinez A et al.
Collagenase-3 (MMP-13) expression by inamed mucosa in inammatory bowel
disease. Scand J Gastroenterol 2006; 41: 10501055.
37 Julovi SM, Ito H, Nishitani K, Jackson CJ, Nakamura T. Hyaluronan inhibits matrix
metalloproteinase-13 in human arthritic chondrocytes via CD44 and P38.
J Orthopaedic Res 2011; 29:258264.
38 Jones CB, Sane DC, Herrington DM. Matrix metalloproteinases: a review of their
structure and role in acute coronary syndrome. Cardiovasc Res 2003; 59: 812823.
39 Zhang X, Chen CT, Bhargava M, Torzilli PA. A. Comparative study of bronectin
cleavage by MMP-1, -3, -13, and -14. Cartilage 2012; 3:267277.
40 Turk BE, Huang LL, Piro ET, Cantley LC. Determination of protease cleavage site motifs
using mixture-based oriented peptide libraries. Nat Biotechnol 2001; 19:661667.
41 Kanda M, Nomoto S, Okamura Y, Hayashi M, Hishida M, Fujii T et al. Promoter
hypermethylation of bulin 1 gene is associated with tumor progression in
hepatocellular carcinoma. Mol Carcinogenesis 2011; 50: 571579.
42 Mikula M, Rubel T, Karczmarski J, Goryca K, Dadlez M, Ostrowski J. Integrating
proteomic and transcriptomic high-throughput surveys for search of new
biomarkers of colon tumors. Funct Integr Genomics 2010.
Calumenin inhibits cell migration by bulin-1
Q Wang et al
12
Oncogene (2014), 1 13 © 2014 Macmillan Publishers Limited
43 Voisin SN, Krakovska O, Matta A, Desouza LV, Romaschin AD, Colgan TJ et al.
Identication of novel molecular targets for endometrial cancer using a drill-down
LC-MS/MS approach with iTRAQ. PLoS One 2010; 6: e16352.
44 Roger P, Pujol P, Lucas A, Baldet P, Rochefort H. Increased immunostaining of
bulin-1, an estrogen-regulated protein in the stroma of human ovarian
epithelial tumors. Am J Pathol 1998; 153:15791588.
45 Pupa SM, Argraves WS, Forti S, Casalini P, Berno V, Agresti R et al. Immunological
and pathobiological roles of bulin-1 in breast cancer. Oncogene 2004; 23:
21532160.
46 Moll F, Katsaros D, Lazennec G, Hellio N, Roger P, Giacalone PL et al.
Estrogen induction and overexpression of bulin-1C mRNA in ovarian cancer cells.
Oncogene 2002; 21: 10971107.
47 Lu J, Guo H, Treekitkarnmongkol W, Li P, Zhang J, Shi B et al. 14-3-3zeta
Cooperates with ErbB2 to promote ductal carcinoma in situ progression to
invasive breast cancer by inducing epithelial-mesenchymal transition. Cancer Cell
2009; 16: 195207.
48 Wang X, Song X, Zhuo W, Fu Y, Shi H, Liang Y et al. The regulatory mechanism of
Hsp90alpha secretion and its function in tumor malignancy. Proc Natl Acad Sci
USA 2009; 106: 2128821293.
49 Lu Y, Xiong Y, Huo Y, Han J, Yang X, Zhang R et al. Grb-2-associated binder 1
(Gab1) regulates postnatal ischemic and VEGF-induced angiogenesis through the
protein kinase A-endothelial NOS pathway. Proc Natl Acad Sci USA 2011; 108:
29572962.
50 Yang M, Baranov E, Jiang P, Sun FX, Li XM, Li L et al. Whole-body optical imaging
of green uorescent protein-expressing tumors and metastases. Proc Natl Acad Sci
USA 2000; 97: 12061211.
51 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.
52 Teng J, Rai T, Tanaka Y, Takei Y, Nakata T, Hirasawa M et al. The KIF3 motor
transports N-cadherin and organizes the developing neuroepithelium. Nat Cell
Biol 2005; 7:474482.
53 Wang Q, Teng J, Shen B, Zhang W, Guo Y, Su X et al. Characterization of kinesin-
like proteins in silkworm posterior silkgland cells. Cell Res 2010; 20: 713727.
54 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;
4:9981008.
55 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:
402408.
Supplementary Information accompanies this paper on the Oncogene website (http://www.nature.com/onc)
Calumenin inhibits cell migration by bulin-1
Q Wang et al
13
© 2014 Macmillan Publishers Limited Oncogene (2014), 1 13
... CALU, encoding reticular calcium-binding proteins, is a low-affinity calcium-binding protein 18 . It is found at extremely low concentrations in the brain 19,20 , which is consistent with our results. Also, the expression of CALU is upregulated in PTE, which might directly regulate calcium channels by directly binding to Ca 2+ through 7 EF-band structures 21 or indirectly regulate calcium channels by manipulating the expressions of glutamic acid (Glu) and aspartic acid 22,23 . ...
Article
Full-text available
The calcium channels are the main pathogenesis and therapeutic target for post-traumatic epilepsy (PTE). However, differentially expressed miRNAs (DEMs) and mRNAs associated with calcium channels in PTE and their interactions are poorly understood. We produced a PTE model in rats and conducted RNA-seq in PTE rats. Gene annotation was used to verify differentially expressed mRNAs related to calcium channels. RNAhybrid, PITA, and Miranda prediction were used to build the miRNA–mRNA pairs. Furthermore, Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were used for the functional enrichment analysis of DEMs. The quantification changes of mRNA and miRNA were verified by RT-qPCR. There were 431 identified differentially expressed genes (DEGs) in PTE rats compared with the sham group, of which five mRNAs and 7 miRNAs were related to calcium channels. The miRNA–mRNA network suggested a negative correlation between 11 pairs of miRNA–mRNA involved in the p53 signaling pathway, HIF-1 signaling pathway. RT-qPCR verified three upregulated mRNAs in PTE rats, associated with 7 DEMs negatively related to them, respectively. This study has revealed the changes in miRNA–mRNA pairs associated with calcium channels in PTE, which might contribute to the further interpretation of potential underlying molecular mechanisms of PTE and the discovery of promising diagnostics.
... Human calumenin is a member of the CREC family and is found throughout the human body, particularly in the endoplasmic reticulum, where it is involved in protein folding and sorting [47]. Further, calumenin inhibits extracellular signal-regulating kinase 1 and 2 (ERK1/2) phosphorylation and cell migration by protecting fibulin-1 from cleavage by MMP-13 [48]. Whether both LPC species inhibit ERK1/2 through calumenin in human FLSs requires further study. ...
Article
Full-text available
Synovial fluid (SF) from human knee joints with osteoarthritis (OA) has elevated levels of lysophosphatidylcholine (LPC) species, but their functional role is not well understood. This in vitro study was designed to test the hypothesis that various LPCs found elevated in OA SF and their metabolites, lysophosphatidic acids (LPAs), modulate the abundance of proteins and phospholipids (PLs) in human fibroblast-like synoviocytes (FLSs), with even minute chemical variations in lysophospholipids determining the extent of regulation. Cultured FLSs (n = 5–7) were treated with one of the LPC species, LPA species, IL-1β, or a vehicle. Tandem mass tag peptide labeling coupled with LC-MS/MS/MS was performed to quantify proteins. The expression of mRNA from regulated proteins was analyzed using RT-PCR. PL synthesis was determined via ESI-MS/MS, and the release of radiolabeled PLs was determined by means of liquid scintillation counting. In total, 3960 proteins were quantified using multiplexed MS, of which 119, 8, and 3 were significantly and reproducibly regulated by IL-1β, LPC 16:0, and LPC 18:0, respectively. LPC 16:0 significantly inhibited the release of PLs and the synthesis of phosphatidylcholine, LPC, and sphingomyelin. Neither LPC metabolite—LPA 16:0 nor LPA 18:0—had any reproducible effect on the levels of each protein. In conclusion, small chemical variations in LPC species can result in the significantly altered expression and secretion of proteins and PLs from FLSs. IL-1β influenced all proteins that were reproducibly regulated by LPC 16:0. LPC species are likely to modulate FLS protein expression only in more advanced OA stages with low IL-1β levels. None of the eight proteins being significantly regulated by LPC 16:0 have been previously reported in OA. However, our in vitro findings show that the CD81 antigen, calumenin, and B4E2C1 are promising candidates for further study, focusing in particular on their potential ability to modulate inflammatory and catabolic mechanisms.
... Thus, this causes calumenin to be distributed throughout the secretory pathway; indeed, it has been found to be secreted from cells [36][37][38]. In addition, calumenin has been found to be located in membranes and interact with various membrane proteins including the ryanodine receptor [39] and cardiac Ca 2+ -transporting ATPase [25], or extracellular matrix proteins such as fibulin [40]. C. elegans has one homolog of calumenin and has been reported to function in various Ca 2+ -regulated behaviors [26]. ...
Article
Full-text available
Caenorhabditis elegans can adapt and survive in dynamically changing environments by the smart and delicate switching of molecular plasticity. C. elegans dauer diapause is a form of phenotypic and developmental plasticity that induces reversible developmental arrest upon environmental cues. An ER (endoplasmic reticulum)-resident Ca2+ binding protein, calumenin has been reported to function in a variety of malignant diseases in vertebrates and in the process of muscle contraction–relaxation. In C. elegans, CALU-1 is known to function in Ca2+-regulated behaviors (pharyngeal pumping and defecation) and cuticle formation. The cuticles of dauer larvae are morphologically distinct from those of larvae that develop in favorable conditions. The structure of the dauer cuticle is thicker and more highly reinforced than that of other larval stages to protect dauer larvae from various environmental insults. Since the calu-1(tm1783) mutant exhibited abnormal cuticle structures such as highly deformed annuli and alae, we investigated whether CALU-1 is involved in dauer formation or not. Ascaroside pheromone (ascr#2) and crude daumone were used under starvation conditions to analyze the rate of dauer formation in the calu-1(tm1783) mutant. Surprisingly, the dauer ratio of the calu-1(tm1783) mutant was extremely low compared to that of the wild type. In fact, the calu-1(tm1783) mutants were mostly unable to enter diapause. We also found that calu-1 is expressed in body-wall muscle and AIA interneurons at the dauer stage. Taken together, our results suggest that CALU-1 is required for normal entry into diapause in C. elegans.
... CALU (Calumenin), as a pivotal gene of the EMT process, has been reported to directly link to the cancer metastasis of a variety of cancers. [43][44][45] CALU may be involved in tumor microenvironment remodeling because of its close association with CD8 cells and macrophages. 46,47 PLOD2 (2-oxoglutarate 5-dioxygenase 2), an intracellular enzyme belonging to the PLOD family, plays a critical role in collagen modification, cell migration, and pulmonary metastasis with no impact on primary tumor growth. ...
Article
Full-text available
Background: Microsatellite instability-high (MSI-H) subgroup of gastric cancer (GC) is characterized by a high tumor mutational burden, increased lymphocytic infiltration, and enhanced inflammatory cytokines. GC patients with MSI-H status have a good response to immune checkpoint blockade management. However, heterogeneity within the subtype and the underlying mechanisms of shaping tumor microenvironments remain poorly understood. Methods: RNA expression levels and clinical parameters of GC were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The data were analyzed using single-sample Gene Set Enrichment Analysis (ssGSEA), univariate Cox regression, multivariate Cox regression, and Least Absolute Shrinkage Selection Operator (LASSO) regression. In addition, multiplex immunohistochemistry (mIHC) was used in our clinical cohort for the tumor microenvironment study. Results: By ssGSEA and survival analysis, the EMT signaling pathway was identified as a representative pathway, which can stratify the patients with MSI-H GC with significant survival predictive power. Then, a novel representative EMT-related five-gene signature (namely CALU, PCOLCE2, PLOD2, SGCD, and THBS2) was established from EMT signaling gene set, which sensitivity and specificity were further validated in the independent GEO database (GSE62254) cohort for disease outcome prediction. Based on public single-cell data and in situ immunohistochemistry, we found that most of these five genes were abundantly expressed in cancer-associated fibroblasts. Furthermore, patients with high or low risk divided by this five-gene signature exhibited a strong correlation of the distinct patterns of tumor immune microenvironment. By mIHC staining of sections from 30 patients with MSI-H status, we showed that the patients with better prognoses had the increased infiltration of CD8+ cells in the primary tumoral tissue. Conclusion: Our study developed a simple five-gene signature for stratifying MSI-H GC patients with survival predictive power.
... With a different approach, an increase in the production of functional rhFIX was demonstrated by Calumenin (CALU) silencing, mediated by small interfering RNA (siRNA) (Wajih et al. 2006). CALU, as a calcium-binding protein, an endoplasmic reticulum chaperone, and a member of an EF-hand superfamily was found to inhibit the γ-carboxylation (Wajih et al. 2004;Wang et al. 2015). Suppression of CALU by shRNA, co-transfected with VKORC1, was also shown to stably enhance the expression of the human coagulation factor VII, in a human cell line (Wajih et al. 2008). ...
Article
Full-text available
Objectives To improve the expression efficiency of recombinant hFIX, by enhancing its γ-carboxylation, which is inhibited by Calumenin (CALU), we used intronic artificial microRNAs (amiRNAs) for the CALU downregulation. Methods Two human CALU (hCALU)-specific amiRNAs were designed, validated and inserted within a truncated form of the hFIX intron 1, in either 3′- or 5′-untranslated regions of the hFIX cDNA, in an expression vector. After transfections of a human cell line with the recombinant constructs, processing of the miRNAs confirmed by RT-PCR, using stem-loop primers. The hFIX and hCALU expression assessments were done based on RT-PCR results. The Gamma(γ)-carboxylation of the expressed hFIX was examined by a barium citrate precipitation method, followed by Enzyme-Linked Immunosorbent Assay. Results Efficient CALU down regulations, with more than 30-fold decrease, occurred in the cells carrying either of the two examined the 3′-located amiRNAs. The CALU downregulation in the same cells doubled the FIX γ-carboxylation, although the transcription of the FIX decreased significantly. On the other hand, while the expression of the amiRNAs from the 5′-located intron had no decreasing effect on the expression level of CALU, the level of hFIX transcription in these cells increased almost twofold compared to the construct without amiRNA. Conclusion The CALU downregulation, consistent with efficient hFIX γ-carboxylation, occurred in the cells carrying either of the two amiRNAs containing constructs, although it was affected by the locations of the amiRNA carrying introns, suggesting a possible need to optimize the conditions for the amiRNAs expression.
... ECM-receptor interactions play an important role in tissue and organ morphogenesis and the maintenance of cell and tissue structure and function. These interactions lead to direct or indirect control of cellular activities such as adhesion, migration, differentiation, proliferation, and apoptosis [60]. By analyzing the enrichment of differential RNA, miRNA and lncRNA and constructing the ceRNA network, we found that the effects of testosterone on backfat adipocytes are more related to adipocyte proliferation, differentiation, migration, apoptosis and signal transduction. ...
Article
Full-text available
Castration can significantly enhance fat deposition in pigs, and the molecular mechanism of fat deposition caused by castration and its influence on fat deposition in different parts of pigs remain unclear. RNA-seq was performed on adipose tissue from different parts of castrated and intact Yorkshire pigs. Different ceRNA networks were constructed for different fat parts. GO and KEGG pathway annotations suggested that testosterone elevates cell migration and affects differentiation and apoptosis in back fat, while it predisposes animals to glycolipid metabolism disorders and increases the expression of inflammatory cytokines in abdominal fat. The interaction between M-7474, novel_miR_243 and SGK1 was verified by dual fluorescence experiments. This ceRNA relationship has also been demonstrated in porcine preadipocytes. Overexpression of M-7474 significantly inhibited the differentiation of preadipocytes compared to the control group. When 100 nM testosterone was added during preadipocyte differentiation, the expression of M-7474 was increased, and preadipocyte differentiation was significantly inhibited. Testosterone can affect preadipocyte differentiation by upregulating the expression of M-7474, sponging novel-miR-243, and regulating the expression of genes such as SGK1. At the same time, HSD11B1 and SLC2A4 may also be regulated by the corresponding lncRNA and miRNA, which ultimately affects glucose uptake by adipocytes and leads to obesity.
... The attached cells were fixed and stained, and cell attachment was quantified. To block specific integrin (integrin β1) or ERK1/2 activity, IPFP-MSCs were pretreated with 10 μg/mL of anti-integrin β1 [33][34][35] (Abcam) or 5 nM EAK1/2 inhibitor SCH772984 [36,37] (TOPSCIENCE) for 12 h, washed with divalent cation-free PBS to removed antibodies or inhibitor and then suspended in TE solution. TE-coated model was also established by adding TE solution to cell-slide, incubated at 37 • C for 2 h, and then washed with double distilled water before cell attachment. ...
Article
Full-text available
Intra-articular injection of mesenchymal stem cells (MSCs) is a promising strategy for osteoarthritis (OA) treatment. However, more and more studies reveal that the injected MSCs have poor adhesion, migration, and survival in the joint cavity. A recent study shows that tropoelastin (TE) regulates adhesion, proliferation and phenotypic maintenance of MSCs as a soluble additive, indicating that TE could promote MSCs-homing in regenerative medicine. In this study, we used TE as injection medium, and compared it with classic media in MSCs intra-articular injection such as normal saline (NS), hyaluronic acid (HA), and platelet-rich plasma (PRP). We found that TE could effectively improve adhesion, migration, chondrogenic differentiation of infrapatellar fat pad MSCs (IPFP-MSCs) and enhance matrix synthesis of osteoarthritic chondrocytes (OACs) in indirect-coculture system. Moreover, TE could significantly enhance IPFP-MSCs adhesion via activation of integrin β1, ERK1/2 and vinculin (VCL) in vitro. In addition, intra-articular injection of TE-IPFP MSCs suspension resulted in a short-term increase in survival rate of IPFP-MSCs and better histology scores of rat joint tissues. Inhibition of integrin β1 or ERK1/2 attenuated the protective effect of TE-IPFP MSCs suspension in vivo. In conclusion, TE promotes performance of IPFP-MSCs and protects knee cartilage from damage in OA through enhancement of cell adhesion and activation of integrin β1/ERK/VCL pathway. Our findings may provide new insights in MSCs intra-articular injection for OA treatment.
Article
Circular RNAs (circRNAs) are involved in tumor growth and metastasis. The aim of this study was to elucidate the possible role of has_circ_0002454 in cervical cancer (CC) and its potential molecular mechanism. has_circ_0002454, miR-654-3p, and SDC4 mRNA levels were determined by RT-qPCR, and SDC4 protein was by Western Blot. Cell proliferation was detected by MTT, colony formation, and wound healing. Invasion and migration capacities of cells were assessed by Transwell assays. The targeting relationship between miR-654-3p and has_circ_0002454 or SDC4 was confirmed by dual-luciferase reporter gene and RIP assay. Xenograft tumor model was constructed to determine the role of has_circ_0002454 in CC. In CC tissues and cell lines, has_circ_0002454 was associated with significantly higher SDC4 levels and significantly lower miR-654-3p expression. Down-regulation of has_circ_0002454 promoted miR-654-3p expression while decreasing SDC4 expression levels. has_circ_0002454 competitively bound miR-654-3p, and elevated miR-654-3p expression rescued the effects of has_circ_0002454 silencing on cancer cell proliferation, apoptosis, migration, and invasion. miR-654-3p directly targeted SDC4, and overexpression of SDC4 reversed the effect of has_circ_0002454 knockdown on cancer cell function. In addition, down-regulation of has_circ_0002454 effectively suppressed tumor growth in vivo. has_circ_0002454 reduction weakens the cancer phenotype of CC cells by modulating the miR-654-3p/SDC4 axis.
Article
Liver hepatocellular carcinoma (LIHC) is a highly lethal form of cancer that is among the deadliest cancer types globally. In terms of cancer‐related mortality rates, liver cancer ranks among the top three, underscoring the severity of this disease. Insufficient analysis has been conducted to fully understand the potential value of the extracellular matrix (ECM) in immune infiltration and the prognostic stratification of LIHC, despite its recognised importance in the development of this disease. The scRNA‐seq data of GSE149614 was used to conduct single‐cell analysis on 10 LIHC samples. CellChat scores were calculated for seven cell populations in the descending cohort to investigate cellular communication, while PROGENy scores were calculated to determine tumour‐associated pathway scores in different cell populations. The pathway analysis using GO and KEGG revealed the enrichment of ECM‐associated genes in the pathway, highlighting the potential role of the ECM in LIHC development. By utilizing the TCGA‐LIHC cohort, an ECM‐based prognostic model for LIHC was developed using Lasso regression. Immune infiltration scores were calculated using two methods, and the performance of the ECM‐related risk score was evaluated using an independent cohort from the CheckMate study. To determine the precise expression of ECM‐associated risk genes in LIHC, we evaluated hepatocellular carcinoma cell lines using a range of assays, including Western blotting, invasion assays and Transwell assays. Using single‐cell transcriptome analysis, we annotated the spatially‐specific distribution of major immune cell types in single‐cell samples of LIHC. The main cell types identified and annotated included hepatocytes, T cells, myeloid cells, epithelial cells, fibroblasts, endothelial cells and B cells. The utilisation of cellchat and PROGENy analyses enabled the investigation and unveiling of signalling interactions, protein functionalities and the prominent influential pathways facilitated by the primary immune cell types within the LIHC. Numerous tumour pathways, including PI2K, EGFR and TGFb, demonstrated a close correlation with the involvement of ECM in LIHC. Moreover, an evaluation was conducted to assess the primary ECM‐related functional changes and biological pathway enrichment in LIHC. Differential genes associated with ECM were identified and utilised to create prognostic models. The prognostic stratification value of these models for LIHC patients was confirmed through validation in multiple databases. Furthermore, through immune infiltration analysis, it was discovered that ECM might be linked to the irregular expression and regulation of numerous immune cells. Additionally, histone acetylation was mapped against gene mutation frequencies and differential expression profiles. The prognostic stratification efficacy of the ECM prediction model constructed in the context of PD‐1 inhibitor therapy was also examined, and it exhibited strong stratification performance. Cellular experiments, including Western blotting, invasion and Transwell assays, revealed that ECM‐associated risk genes have a promoting effect on the development of LIHC. The creation of biomarkers for LIHC using ECM‐related genes unveiled substantial correlations with immune microenvironmental infiltration and functional mutations in various tumour pathways. This enlightens us to the possibility that the influence of ECM on tumours may extend beyond simply promoting the fibrotic process and the stromal composition of tumours.
Article
Human calumenin-1 (HsCalu-1) is an endoplasmic reticulum (ER) and Golgi-resident Ca2+-binding protein of the hepta-EF-hand superfamily that plays a vital role in maintaining the cytoplasmic Ca2+ concentration below toxic levels by interacting with Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) and ryanodine receptors (RyR), indicating its role in Ca2+ homeostasis in the ER. HsCalu-1 seems to be able to exhibit structural plasticity to achieve its plethora of functions. In this study, we demonstrate that HsCalu-1 acts as a chaperone in both its intrinsically disordered state (apo form) and the structured state (Ca2+-bound form). HsCalu-1 chaperone activity is independent of Ca2+ and Pb2+ binding attenuating its chaperone-like activity. Incidentally, Pb2+ binds to HsCalu-1 with lower affinity (KD = 38.46 μM) (compared to Ca2+-binding), leading to the formation of a less-stable conformation as observed by a sharp drop in its melting temperature Tm from 67 °C in the Ca2+-bound form to 43 °C in the presence of Pb2+. The binding site for Pb2+ was mapped as being in the EF-Hand-234 domain of HsCalu-1, a region that overlaps with the Ca2+-dependent initiator of its functional fold. A change in the secondary and tertiary structure, leading to a less-stable but compact conformation upon Pb2+ binding, is the mechanism by which the chaperone-like activity of HsCalu-1 is diminished. Our results not only demonstrate the chaperone activity by a protein in its disordered state but also explain, using Pb2+ as a probe, that the multiple functions of calumenin are due to its ability to adopt a quasi-stable conformation.
Article
Full-text available
The ability of cells to adhere to each other and to their surrounding extracellular matrices is essential for a multicellular existence. Adhesion provides physical support for cells, regulates cell positioning and enables microenvironmental sensing. The integrins and the syndecans are two adhesion receptor families that mediate adhesion, but their relative and functional contributions to cell-extracellular matrix interactions remain obscure. Recent advances have highlighted connections between the signalling networks that are controlled by these families of receptors. Here we survey the evidence that synergistic signalling is involved in controlling adhesive function and the regulation of cell behaviour in response to the external environment.
Article
Full-text available
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.
Article
Full-text available
Fibronectin fragments are important for synovial inflammation and the progression of arthritis, and thus, identifying potential enzymatic pathways that generate these fragments is of vital importance. The objective of this study was to determine the cleavage efficiency of fibronectin by matrix metalloproteinases (MMP-1, MMP-3, MMP-13, and MMP-14). Intact human plasma fibronectin was co-incubated with activated MMPs in neutral or acidic pH for up to 24 hours at 37 °C. The size and distribution of fibronectin fragments were determined by Western blot analysis using antibodies that recognized the N-terminals of fibronectin. All MMPs were able to cleave fibronectin at neutral pH. MMP-13 and -14 had the highest efficiency followed by MMP-3 and -1. MMP-3, -13, and -14 generated 70-kDa fragments, a known pro-inflammatory peptide. Further degradation of fibronectin fragments was only found for MMP-13 and -14, generating 52-kDa, 40-kDa, 32-kDa, and 29-kDa fragments. Fibronectin fragments of similar size were also found in the articular cartilage of femoral condyles of normal bovine knee joints. At acidic pH (5.5), the activities of MMP-1 and -14 were nearly abolished, while MMP-3 had a greater efficiency than MMP-13 even though the activities of both MMPs were significantly reduced. These findings suggest that MMP-13 and -14 may play a significant role in the cleavage of fibronectin and the production of fibronectin fragments in normal and arthritic joints.
Article
Full-text available
Several pathological processes, such as sepsis and inflammatory bowel disease (IBD), are associated with impairment of intestinal epithelial barrier. Here, we investigated the role of matrix metalloproteinase MMP13 in these diseases. We observed that MMP13(-/-) mice display a strong protection in LPS- and caecal ligation and puncture-induced sepsis. We could attribute this protection to reduced LPS-induced goblet cell depletion, endoplasmic reticulum stress, permeability and tight junction destabilization in the gut of MMP13(-/-) mice compared to MMP13(+/+) mice. Both in vitro and in vivo, we found that MMP13 is able to cleave pro-TNF into bioactive TNF. By LC-MS/MS, we identified three MMP13 cleavage sites, which proves that MMP13 is an alternative TNF sheddase next to the TNF converting enzyme TACE. Similarly, we found that the same mechanism was responsible for the observed protection of the MMP13(-/-) mice in a mouse model of DSS-induced colitis. We identified MMP13 as an important mediator in sepsis and IBD via the shedding of TNF. Hence, we propose MMP13 as a novel drug target for diseases in which damage to the gut is essential.
Article
Full-text available
Precise spatiotemporal coordination of integrin adhesion complex dynamics is essential for efficient cell migration. For cells adherent to fibronectin, differential engagement of α5β1 and αVβ3 integrins is used to elicit changes in adhesion complex stability, mechanosensation, matrix assembly, and migration, but the mechanisms responsible for receptor regulation have remained largely obscure. We identify phosphorylation of the membrane-intercalated proteoglycan syndecan-4 as an essential switch controlling integrin recycling. Src phosphorylates syndecan-4 and, by driving syntenin binding, leads to suppression of Arf6 activity and recycling of αVβ3 to the plasma membrane at the expense of α5β1. The resultant elevation in αVβ3 engagement promotes stabilization of focal adhesions. Conversely, abrogation of syndecan-4 phosphorylation drives surface expression of α5β1, destabilizes adhesion complexes, and disrupts cell migration. These data identify the dynamic spatiotemporal regulation of Src-mediated syndecan-4 phosphorylation as an essential switch controlling integrin trafficking and adhesion dynamics to promote efficient cell migration.
Article
Full-text available
Aggrecan, versican, neurocan, and brevican are important components of the extracellular matrix in various tissues. Their amino-terminal globular domains bind to hyaluronan, but the function of their carboxyl-terminal globular domains has long remained elusive. A picture is now emerging where the C-type lectin motif of this domain mediates binding to other extracellular matrix proteins. We here demonstrate that aggrecan, versican, and brevican lectin domains bind fibulin-2, whereas neurocan does not. As expected for a C-type lectin, the interactions are calcium-dependent, withK D values in the nanomolar range as measured by surface plasmon resonance. Solid phase competition assays with previously identified ligands demonstrated that fibulin-2 and tenascin-R bind the same site on the proteoglycan lectin domains. Fibulin-1 has affinity for the common site on versican but may bind to a different site on the aggrecan lectin domain. By using deletion mutants, the interaction sites for aggrecan and versican lectin domains were mapped to epidermal growth factor-like repeats in domain II of fibulin-2. Affinity chromatography and solid phase assays confirmed that also native full-length aggrecan and versican bind the lectin domain ligands. Electron microscopy confirmed the mapping and demonstrated that hyaluronan-aggrecan complexes can be cross-linked by the fibulins.
Article
Gelatinase A and membrane-type metalloproteinase (MT1-MMP) were able to process human procollagenase 3 (M(r) 60,000) to the fully active enzyme (Tyr(85) N terminus; M(r) 48,000). MT1-MMP activated procollagenase-3 via a M(r) 56,000 intermediate (Ile(36) N terminus) to 48,000 which was the result of the cleavage of the Glu(84)-Tyr(85) peptide bond. We have established that the activation rate of procollagenase-3 by MT1-MMP was enhanced in the presence of progelatinase A, thereby demonstrating a unique new activation cascade consisting of three members of the matrix metalloproteinase family. In addition, procollagenase-3 can be activated by plasmin, which cleaved the Lys(38)-GlU(39) and Arg(76)-Cys(77) peptide bonds in the propeptide domain, Autoproteolysis then resulted in the release of the rest of She propeptide domain generating Tyr(85) N-terminal active collagenase 3. However, plasmin cleaved the C-terminal domain of collagenase-3 which results in the loss of its collagenolytic activity. Concanavalin A-stimulated fibroblasts expressing MT1-MMP and fibroblast-derived plasma membranes were able to process human procollagenase-3 via a M, 56,000 intermediate form to the final M(r) 48,000 active enzyme which, by analogy with progelatinase A activation, may represent a model system for in vivo activation. Inhibition experiments using tissue inhibitor of metalloproteinases, plasminogen activator inhibitor-a, or aprotinin demonstrated that activation in the cellular model system was due to MT1-MMP/gelatinase A and excluded the participation of serine proteinases such as plasmin during procollagenase-3 activation, We have established that progelatinase A can considerably potentiate the activation rate of procollagenase-3 by crude plasma membrane preparations from concanavalin A-stimulated fibroblasts, thus confirming our results using purified progelatinase A and MT1-MMP. This new activation cascade may be significant in human breast cancer pathology, where all three enzymes have been implicated as playing important roles.
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.