ArticlePDF Available

New facets of matrix metalloproteinases MMP-2 and MMP-9 as cell surface transducers: Outside-in signaling and relationship to tumor progression

Authors:

Abstract and Figures

This review focuses on matrix metalloproteinases (MMPs)-2 (gelatinase A) and-9 (gelatinase B), both of which are cancer-associated, secreted, zinc-dependent endopeptidases. Gelatinases cleave many different targets (extracellular matrix, cytokines, growth factors, chemokines and cytokine/growth factor receptors) that in turn regulate key signaling pathways in cell growth, migration, invasion, inflammation and angiogen-esis. Interactions with cell surface integral membrane proteins (CD44, αVβ/αβ1/αβ2 integrins and Ku protein) can occur through the gelatinases' active site or hemopexin-like C-terminal domain. This review evaluates the recent literature on the non-enzymatic, signal transduction roles of surface-bound gelatinases and their subsequent effects on cell survival, migration and angiogenesis. Gelatinases have long been drug targets. The current status of gelatinase inhibitors as anticancer agents and their failure in the clinic is discussed in light of these new data on the gelatinases' roles as cell surface transducers — data that may lead to the design and development of novel, gelatinase-targeting inhibitors.
Content may be subject to copyright.
Review
New facets of matrix metalloproteinases MMP-2 and MMP-9 as cell surface
transducers: Outside-in signaling and relationship to tumor progression
Brigitte Bauvois
INSERM U872, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Université Paris Descartes, Paris, France
abstractarticle info
Article history:
Received 15 June 2011
Received in revised form 3 October 2011
Accepted 4 October 2011
Available online 12 October 2011
Keywords:
Gelatinase
Cell surface binding
Cancer
Inhibitor
Function
Outside-in signaling
This review focuses on matrix metalloproteinases (MMPs)-2 (gelatinase A) and -9 (gelatinase B), both of
which are cancer-associated, secreted, zinc-dependent endopeptidases. Gelatinases cleave many different
targets (extracellular matrix, cytokines, growth factors, chemokines and cytokine/growth factor receptors)
that in turn regulate key signaling pathways in cell growth, migration, invasion, inammation and angiogen-
esis. Interactions with cell surface integral membrane proteins (CD44, αVβ/αβ1/αβ2 integrins and Ku pro-
tein) can occur through the gelatinases' active site or hemopexin-like C-terminal domain. This review
evaluates the recent literature on the non-enzymatic, signal transduction roles of surface-bound gelatinases
and their subsequent effects on cell survival, migration and angiogenesis. Gelatinases have long been drug
targets. The current status of gelatinase inhibitors as anticancer agents and their failure in the clinic is dis-
cussed in light of these new data on the gelatinases' roles as cell surface transducers data that may lead
to the design and development of novel, gelatinase-targeting inhibitors.
© 2011 Elsevier B.V. All rights reserved.
Contents
1. Introducing MMPs and their roles in cancer ................................................ 29
2. Development of synthetic MMP inhibitors as cancer drugs ......................................... 31
3. Gelatinases (MMP-2 and MMP-9) as cancer biomarkers........................................... 31
4. Proteolysis-dependent functions of gelatinases ............................................... 31
5. Cell surface-associated gelatinases..................................................... 32
6. Outside-in signaling by cell surface-associated gelatinases ......................................... 33
7. Open questions and conclusions...................................................... 33
Acknowledgement .............................................................. 34
References .................................................................. 35
1. Introducing MMPs and their roles in cancer
The matrix metalloproteinase (MMP) family consists of at least 23
structurally related, zinc-dependent endopeptidases [1,2]. The family
shares specic functional and structural components, including a
hydrophobic signal peptide for secretion, a propeptide domain for
enzyme latency, a catalytic domain with a highly conserved zinc-
binding site and (for the majority of MMPs) a hemopexin-like
C-terminal domain (PEX) linked to the catalytic domain via aexible
hinge region (Fig. 1A) [1,2]. The PEX domain binds endogenous tissue
inhibitors of MMPs (TIMPs) and certain MMP substrates and is in-
volved in MMP activation [1]. TIMPs include four members originally
described as inhibitors of MMP activities, which also have biological ac-
tivities that are independent of MMP inhibition and regulate cell growth,
migration, survival and angiogenesis [35]. MMPs include membrane-
Biochimica et Biophysica Acta 1825 (2012) 2936
Abbreviations: MMP, matrix metalloproteinase; PEX, hemopexin-like C-terminal
domain; TIMP, tissue inhibitor of metalloproteinase; MT, membrane type; CBD,
collagen-binding domain; ECM, extracellular matrix; MMPI, matrix metalloproteinase
inhibitor; CLL, chronic lymphocytic leukemia; AML, acute myeloid leukemia; EGF,
epidermal growth factor; VEGF, vascular endothelial growth factor; TGF-β, transforming
growth factor-β;FGF,broblast growth factor; TNF, tumor necrosisfactor; IL, interleukin;
IFN, interferon; IGF-BP, insulin-like growth factor-binding protein; ICAM, intercellular
adhesion molecule; PF4, platelet factor-4; SDF-1, stromal-cell derived factor-1; IP-10,
IFN-γ-induced protein of 10 kDa; MCP, macrophage chemotactic protein; I-TAC, IFN-γ-
induced T cell-activated chemokine; MIG, monokine induced by interferon-γ;RECK,
reversion-inducing cysteine-rich protein with Kazal motif; LRP, low density lipoprotein-
related scavenger receptor; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated
protein kinase; ERK, extracellular signal-regulated protein kinase; FAK, focal adhesion
kinase; CAM, chick chorioallantoic membrane; HIF, hypoxia-induced transcription factor
Centre de Recherche des Cordeliers, INSERM U872, 15 rue de l'Ecole de Médecine,
F-75270 Paris cedex 06, France. Tel.: +33 144 278 188; fax: + 33 144 278 161.
E-mail address: brigitte.bauvois@crc.jussieu.fr.
4
0304-419X/$ see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.bbcan.2011.10.001
Contents lists available at SciVerse ScienceDirect
Biochimica et Biophysica Acta
journal homepage: www.elsevier.com/locate/bbacan
anchored and secreted MMPs. The membrane-anchored MMPs (MT-
MMPs) are localized at the cell surface by a C-terminal (type I) trans-
membrane domain or a glycosylphosphatidylinositol anchor (Fig. 1B)
[4,6]. A type II transmembrane MMP (MMP-23) has also been described
[6]. TIMP-1 has a relatively low afnity for the MT-MMPs [5].MMPsse-
creted as latent pro-enzymes include collagenases, stromelysins, matrily-
sins and two gelatinases (A and B) (Fig. 1C) [4].Removalofthe
prodomain by the endopeptidase furin leads to MMP activation. TIMPs
inhibit most of the secreted MMPs [5]. The gelatinases differ from most
of the other MMPs in that they have a collagen-binding domain (CBD)
within the catalytic domain (Fig. 1C). The CBD is composed of three
bronectin type II repeats and is involved in the binding of collagenous
substrates, elastin, fatty acids and thrombospondins [7].
Matrix metalloproteinases selectively degrade various compo-
nents of the extracellular matrix (ECM) and release growth factors
and cytokines that reside in the ECM [8,9]. The MMPs are also capa-
ble of activating various latent growth factors, cytokines and chemo-
kines and cleaving cell surface proteins (cytokine receptors, cell
adhesion molecules, the urokinase receptor, etc.) [1,2,10,11].
Through their proteolytic activity, MMPs play crucial roles in inva-
sion and metastasis and regulate signaling pathways that control
cell growth, survival, invasion, inammation and angiogenesis
hinge
linker
Pre Pro catalytic Zn2+
SS
PEX
(A) Basic MMP structure
(C) Secreted matrilysins
collagenases,
stromelysins
gelatinases
A & B
SS
SS
CBD
(B) Membrane-anchored
SS
GPI
TM-I
cytoplasmic
tail
SS
Fig. 1. Structures of the MMPs. (A) The general domain structure of MMP family members. The signal peptide (Pre) guides the MMP into the rough endoplasmic reticulum during
synthesis. The propeptide domain (Pro) sustains the latency of MMPs. The catalytic domain houses a highly conserved Zn
2+
binding region. The hemopexin-like-C-terminal domain
(PEX) is linked to the catalytic domain by a short hinge region. (B) MT-MMPs include membrane-anchored MMPs localized at the cell surface through a C-terminal (type I) trans-
membrane domain (TM-I) or by a glycosylphosphatidylinositol anchor (GPI). (C) Secreted MMPs include stromelysins, matrilysins, collagenases and gelatinases. The gelatinases
(MMP-2 and MMP-9) contain repeats of bronectin type II-like domains (the collagen binding domain, CBD) that interact with collagen and gelatin.
MMPs
TIMPs
Proteolysis of ECM
Release of:
growth factors
angiogenic factors
Processing of:
cytokines
chemokines
growth factors
growth factor binding proteins
cell adhesion molecules
receptors
Growth Survival
Invasion Metastasis
Angiogenesis
Inflammation
endothelial,
inflammatory
and tumor cells
Fig. 2. A schematic overview of the roles of MMPs in cancer. MMPs degrade structural components within the ECM, facilitating tumor cell invasion and metastasis and thus releasing
growth factors, cytokines and angiogenic factors embedded in the ECM (VEGF, TGF-β, bFGF, IFN-γ, etc.). MMPs also generate angiogenesis inhibitors, such as angiostatin, endostatin
and tumstatin. MMPs process and activate or inactivate signaling molecules (cytokines, chemokines, growth factors) that target immune cells (inammation), endothelial cells (an-
giogenesis) and tumor cells (cell growth, survival, migration, invasion and metastasis). MMP-mediated cleaving of adhesion molecules (E-cadherin, ICAM-1, integrins, etc.)
enhances tumor cell migration and invasion. ( negative regulation, positive regulation).
30 B. Bauvois / Biochimica et Biophysica Acta 1825 (2012) 2936
(Fig. 2). A number of excellent reviews have discussed the MMPs'
roles in cancer [1,2,8,1214].
2. Development of synthetic MMP inhibitors as cancer drugs
Many different MMP inhibitors (MMPIs) have been designed to
target MMPs in cancer [13,15,16]. Although these compounds differ
in their inhibitory potencies towards MMPs, none of them are selec-
tive for a given MMP (including the gelatinases).
The rst generation of MMPIs were peptidometics (such as bati-
mastat and marimastat) that mimic the structure of collagen. They
act as competitive inhibitors and chelate the zinc ion present at the
MMP's active site. To improve specicity and oral bioavailability,
non-peptidometics (such as tanomastat, prinomastat, BMS-275291,
CGS27123A, etc.) were synthesized on the basis of the active site's
three-dimensional conformation. Other MMPIs include tetracycline
derivatives (such as metastat/COL-3, minocycline and doxycycline)
that inhibit both the MMPs' enzymatic activity and their synthesis
(by blocking gene transcription) [17,18]. To date, all clinical trials of
these MMPIs in advanced cancer patients have failed, with the excep-
tion of metastat (which has entered Phase II trials for Kaposi's
sarcoma and brain tumors) [19,20]. The latest generation of MMPIs
includes biphosphonates [21,22] and S-3304, a D-tryptophan deriva-
tive that primarily inhibits gelatinases [23]. Novel biphosphonate de-
rivatives show benets as a result of altering the expression pattern of
MMPs/TIMPs in breast cancer cells [13]. A Phase I clinical trial of
S-3304 in patients with advanced and refractory solid tumors found
that the compound was safe, well tolerated and achieved plasma con-
centrations above those required to inhibit gelatinases [24]. However,
it is not yet known whether S-3340 will be effective in Phase II/III
clinical trials.
There are several possible reasons for the failure of MMPIs in the
clinic. Firstly, most MMPIs have dose-limiting musculoskeletal toxici-
ty that limits efcacy. Secondly, the clinical trials were performed on
patients with terminal-phase cancer, where several overlapping
pathways come into play. Thirdly, the structural similarity of the var-
ious MMPs' catalytic domains makes it difcult to design MMPIs with
high selectivity [25]. Moreover, the role of MMPs in cancer progres-
sion appears not to be restricted to their ECM-degrading activity,
with involvement in many signaling pathways that inuence tumor
cell behavior [26,27]. Lastly, recent evidence shows that MMPs may
have opposing functions in primary and metastatic cancer sites;
hence, MMPIs may produce protumorigenic effects in some situations
and may counterbalance the benets of target inhibition [26,28,29].
3. Gelatinases (MMP-2 and MMP-9) as cancer biomarkers
Of the various MMPs thought to be involved in cancer, attention
has focused on the gelatinases because (i) they are overexpressed in
a variety of malignant tumors and (ii) their expression and activity
are often associated with tumor aggressiveness and a poor prognosis.
Elevated levels of MMP-2 and/or MMP-9 are found in breast, brain,
ovarian, pancreas, colorectal, bladder, prostate and lung cancers and
melanoma [2,8,3032]. Dysregulated MMP expression is also ob-
served in hematological malignancies such as acute lymphoblastic
leukemia, adult T-cell leukemia, chronic B lymphocytic leukemia
(CLL), acute myeloid leukemia (AML), chronic myeloid leukemia,
myelodysplastic syndromes and Hodgkin's and non-Hodgkin's lym-
phoma [31,33].
4. Proteolysis-dependent functions of gelatinases
Gelatinases are secreted as inactive zymogens (proMMP-2:
72 kDa, proMMP-9: 92 kDa), with cleavage of a prodomain yielding
the active form (MMP-2: 65 kDa; MMP-9: 82 kDa). An 85 kDa pro-
form of MMP-9 lacking complex carbohydrates has been reported in
breast tumors and AML cell lines [34,35]. Several mechanisms can
stimulate the activation process. The main route for activation of
proMMP-2 on the cell surface occurs through the formation of a mo-
lecular complex containing proMMP-2 (via its PEX domain), MT1-
MMP (via its catalytic domain) and TIMP-2 (reviewed in [2]). This
cell surface interaction leads to clustering of proMMP-2 near a
TIMP-free, active MT1-MMP which initiates activation of proMMP-2.
MMP-2 can also be activated by MMP-1, MMP-7, thrombin and acti-
vated protein C [7,36]. MMP-9 can be activated by plasmin, trypsin-
2, MMP-2, MMP-13 (activated by MMP-2) and MMP-3 (activated by
plasmin) [7,36]. Other activation mechanisms have been suggested
in order to explain proMMP-2 and proMMP-9's catalytic activity in
the presence of the propeptide. For example, binding of proMMP-9
to a gelatin- or type IV collagen-coated surface could lead to revers-
ible activation of MMP-9 via disengagement of the propeptide from
the active site [37]. Interaction of hemin or β-hematin with the
proMMP-9 PEX domain primes MMP-9 activation via an autocatalytic
process [38]. Interaction of proMMP-2 with low concentrations of col-
lagen α2 VI chain induces its auto-activation [39]. Lastly, the
reversion-inducing cysteine-rich protein with Kazal motifs (RECK,
an integral membrane protein that forms a complex with MT1-
MMP) has been found to inhibit gelatinase secretion and activity
[40,41]. Whether these mechanisms occur in vivo remains to be
established.
Activated gelatinases are able to degrade various components of
the ECM and non-matrix proteins (Table 1)[1,2,8,11,12]. Cell migra-
tion and invasion are complex processes that involve the ECM, pro-
teinases, chemokines, adhesion receptors and (for invasion)
basement membrane. Angiogenesis (dened as the generation of
new blood vessels from preexisting ones) is critically important for
tumor growth and metastatic spreading. It was initially suggested
that gelatinases played a dominant role in basement membrane-
invasive events because of their ability to degrade collagen IV [42].
However, studies in MMP-9
(/)
and MMP-2
(/)
/MMP-9
(/)
mu-
rine models of inammation [43], cancer cells engineered to express
active MMP-2 and MMP-9 [44] and broblasts isolated from MMP-
2
/
and MMP-9
/
mice [45] strongly suggest that gelatinases do
not promote basement membrane invasion. In fact, recent evidence
shows that gelatinases play major but indirect roles in cell signaling
by controlling the bioavailability and bioactivity of molecules that tar-
get specic receptors regulating cell growth, migration, inammation
and angiogenesis (Table 1).
Table 1
Gelatinase substrates.
Substrates MMP-2/gelatinase A MMP-9/gelatinase B
ECM
substrates
Collagens I, IV, V, VII, X and XI Collagens III, IV and V
Gelatin Gelatin
Tenascin Elastin
Elastin Vitronectin
Fibronectin Entactin
Laminin-5
Other
substrates
proTGF-βproTGF-β
proIL-1βproTNF-α
proTNF-αIL-2Rα
proHB-EGF ICAM-1
FGFR-I EGFR-1
IGFBP-3, -5, -6 Kit ligand
CXCL12/SDF-1 CXCL1/GRO-α
CCL7/MCP-3 CXCL4/PF4
CX3CL1/fractalkine CXCL8/IL-8
KISS-1 CXCL9/MIG
CXCL11/ITAC
CXCL12/SDF-1
α1 proteinase inhibitor
Plasminogen
KISS-1
IFN-β
31B. Bauvois / Biochimica et Biophysica Acta 1825 (2012) 2936
By degrading the ECM, gelatinases generate or release bioactive
molecules that inuence tumor progression. Gelatinase activity can
cause the release of cryptic information from the ECM, leading to
cell migration and angiogenesis. For example, the proteolytic cleav-
age of collagen IV by MMP-9 unmasks cryptic sites that are critical
for angiogenesis [46,47]. Similarly, cleavage of laminin-5 by MMP-2
results in the exposure of a cryptic epitope that enhances endothelial
cell migration [48]. MMP-9 can release ECM-sequestered factors
VEGF, TGF-βand FGF-2, which stimulate proliferation and migration
of endothelial cells and thus promote angiogenesis and tumor growth
[4953]. In contrast, tumstatin and endostatin (generated by the
MMP-9-mediated proteolysis of type IV collagen and type XVIII colla-
gen, respectively) are active inhibitors of angiogenesis [54,55].
Gelatinases target immunomodulating cytokines and growth fac-
tors and cytokine/growth factor receptors. For example, gelatinases
shed and activate TNF-α, TGF-βand IL-1β, which are intimately in-
volved in the regulation of growth, angiogenesis and inammation
[56,57]. FGF-R1 may be a specic cell-surface target for MMP-2, yield-
ing a soluble FGF receptor that modulates the mitogenic and angio-
genic activities of FGF-2 [58]. MMP-9 cleaves IFN-βand thus kills
the cytokine's antiviral and immunotherapeutic activity [59]. MMP-
2 is able to cleave certain insulin-like growth factor-binding proteins
(IGFBPs) and thus release active insulin-like growth factors (IGFs) in-
volved in tumor cell growth [11,60]. Both gelatinases process the
tumor suppressor protein KISS-1 to generate metastin, which en-
hances cell invasion [61,62]. MMP-9 suppresses the proliferation of
T lymphocytes through disruption of the IL-2Rαsignaling that may
constitute a mechanism of cancer-mediated immunosuppression
[63]. Moreover, MMP-9 releases Kit-ligand, which plays a crucial
role in tumor growth and angiogenesis [64,65]. MMP-9-dependent
shedding of ICAM-1 augments tumor cell resistance to natural-
killer-cell-mediated cytotoxicity [66].
Chemokines play an essential role in modulating tumor growth via
regulation of tumor-associated angiogenesis, activation of host immu-
nological responses and direct inhibition of tumor cell proliferation.
Gelatinases generate either inactivated chemokine fragments (e.g.
GRO-α, PF4, SDF-1, MCP-3, IP-10, MIG) or truncated chemokines
with enhanced activity (IL-8, I-TAC) [9]. The gelatinase-mediated pro-
teolysis of chemokines might have direct consequences on tumor
growth (e.g. I-TAC), migration (e.g. SDF-1 and MCP-3) and angiogene-
sis (e.g. IL-8, PF4, MIG, IP-10 and SDF-1) [7,9,11]). For example, the
MCP-3 generated by MMP-2 can bind to CC chemokine receptors
and inhibit migration, and suppresses inammation [67]. In contrast,
processing of IL-8 by MMP-9 increases its chemotactic activity in neu-
trophils [68].
5. Cell surface-associated gelatinases
Gelatinases have been shown to interact with the cell surfaces of
leukocytes and epithelial and endothelial cells [7,36,69,70]. As men-
tioned above, the activation of proMMP-2 requires interaction with
MT1-MMP and TIMP-2 [7]. Furthermore, gelatinases bind to collagens
and bronectin at the surface of cancer cells through their CBD do-
main [7]. Gelatinases also bind to the low-density lipoprotein-
related scavenger receptor (LRP), which is responsible for the inter-
nalization of various ligands including these enzymes [7173].
In addition to these cell surface associations, gelatinases reported-
ly bind to other integral membrane proteins, such as the DNA repair
protein Ku (via its integrin I-like domain) [74], CD44 [50,7580] and
the integrins (αVβ3, αVβ1, αβ2, αVβ5, α4β1 and α5β1) [70,8093]
(Table 2). The gelatinases' catalytic and PEX domains are variously in-
volved in these interactions (Table 2). For example, the integrin α
M
,
α
L
and β2 subunits can bind to MMP-9's catalytic domain, whereas
CD44 and the β5, α4 and β1 subunits interact with the PEX domain
(Table 2). However, doubt has been cast on the reported molecular
interaction between MMP-2 and integrin αVβ3via the PEX domain
in endothelial cells [82], with a suggestion that the recombinant
PEX polypeptide was possibly contaminated by lipopolysaccharide
[94]. On mesenchymal cells, active MMP-2 can bind to αVβ3via
Table 2
Examples of binding between gelatinases and integral membrane proteins.
Cell type Gelatinase (domain involved in the binding) Integral membrane protein Refs Positive effect on cell process(es)
Melanoma
Primary and metastatic melanomas active MMP-2 αVβ3[81,85] Growth collagen IV degradation
MC cells pro/active MMP-9 CD44 [76] Growth migration
1
Squamous cell carcinoma
SCC12F2 cells pro/active MMP-9 α5β1[88] Migration
2
Breast cancer
MCF7, MCF10A cells pro/active MMP-2 αVβ3[82,83 Migration angiogenesis
3
Met-1, MDA-MB435 cells active MMP-9 αVβ3[75,87 Migration
4, 5
MMP-9 transfected MCF-7 cells proMMP-9 (PEX) CD44 [78,79 Migration
5
Lung cancer
A549 cells MMP-2 (catalytic) αVβ3[93] VEGF release and angiogenesis
6
Fibrosarcoma
HT1080 cells proMMP-9 (PEX) αVβ5[89] Migration
5,7
AML
THP-1, OCI-AML3 cells pro MMP-2 (catalytic) α
M
β2, α
L
β2[90] migration
5
THP-1 cells proMMP-9 (catalytic) α
M
β2, α
L
β2[90,91] Growth migration
5
transendothelial migration
8
U937, HL-60, THP-1, primary AML blasts proMMP-9 (PEX) Ku protein [35,74] Migration
9
CLL
Primary CLL cells pro/active MMP-9 (PEX) α4β1 and CD44v [80,96] Survival
Leukocytes Migration
5
Neutrophils, monocytes proMMP2 and proMMP-9 (catalytic) α
L
β2, α
M
β2[91] Transendothelial migration
8
Monocyte-derived dendritic cells active MMP-9 (catalytic) α
M
β2 and CD44 [77] Migration
10
Endothelial cells
ECV304 cells proMMP-2 αVβ3[82,85] Growth angiogenesis
3
HUVEC cells active MMP-2 αVβ1[86] Apoptosis
1
In vitro culture of TA3 cells on G8 myoblast monolayers; in vitro endothelial tube formation;
2
cell migration in bronectin-coated Transwell
®
chambers;
3
CAM angiogenesis assay;
4
cell migration in puried ECM proteins-coated Transwell
®
chambers;
5
cell migration in Transwell
®
chambers with 10% fetal calf serum as chemoattractant in the lower chamber;
6
in vitro culture of HMEC-1 cells on Matrigel
®
and formation of capillary-like structures; xenograft mice;
7
cell migration in Matrigel
®
-coated Transwell
®
chambers;
8
transendothe-
lial cell migration in HMECs-coated Transwell
®
chambers;
9
cell migration in collagen IV-coated Transwell
®
chambers;
10
cell migration in Matrigel
®
-coated Transwell
®
chambers
with chemokine in the lower chamber.
32 B. Bauvois / Biochimica et Biophysica Acta 1825 (2012) 2936
PEX [92]. It therefore remains to be denitively established whether
αVβ3 can bind to the PEX domain of MMP-2.
Consequently, cell growth, migration and angiogenesis appear to
depend on cell surface associations between gelatinases and these in-
tegral membrane proteins (Table 2). For example, disrupting MMP-2/
αvβ3 binding on the surface of melanoma cells is associated with the
inhibition of tumor growth and migration [84,95]. Similarly, the com-
plex formed by Ku protein and proMMP-9 (via the latter's PEX do-
main) is involved in the migration of AML cells [35]. Both pro-
MMP-9 and active MMP-9 bound to the membrane via α4β1 and
CD44 are involved in the survival of CLL cells [80,96]. ProMMP-9 en-
hances epithelial cell migration via a non-proteolytic mechanism
that involves its PEX domain and CD44 [78]. Mesenchymal invasive
behavior might be dependent on MMP-2/αVβ3 binding [92]. In neu-
trophils, both the active site and PEX domain of MMP-9 are involved
in the induction of FGF-2-mediated angiogenesis [53].
6. Outside-in signaling by cell surface-associated gelatinases
Observation of these binding associations between surface recep-
tors and gelatinases raised the possibility that the latter have the po-
tential to directly inuence cell behavior and to activate the classical
signaling pathways involved in major biological events (cell growth,
migration, survival, etc.).
Experiments with inhibitors strongly suggest the involvement of
cell signaling pathways in MMP-9-mediated cell migration. For exam-
ple, the JNK inhibitor SP600125 blocked MMP-9-mediated dendritic
cell migration [97], whereas the MAPK inhibitor PD98059 and the
PI3K inhibitor LY-294002 inhibited MMP-9-induced epithelial cell
migration [78]. By studying the adenovirus-mediated delivery of
MMP small interfering RNA, Rao, Bhoopathi and colleagues showed
a clear relationship between the loss of MMP-9 expression and apo-
ptosis induction in medulloblastoma cells (associated with the activa-
tion of β1 integrin, ERK and NF-κB) [98,99].
By using a combination of strategies to respectively target MMPs
(with siRNA, recombinant MMPs and enzyme inhibitors),
gelatinase-integral protein interactions (with antibodies) and signal
transduction pathways (with signaling inhibitors and siRNA), three
recent studies have described the signaling properties of MMP-2
and MMP-9. Redondo-Munoz and colleagues showed that the bind-
ing of proMMP-9's PEX domain to its docking receptors α4β1 integrin
and CD44 induces an intracellular signaling pathway that favors the
survival of CLL cells [96]. This pathway (Fig. 3A) consists of Lyn kinase
activation, STAT3 phosphorylation and up-regulated expression of
the pro-survival protein Mcl-1 (a member of the Bcl-2 family). Ac-
cordingly, high levels of proMMP-9 and Mcl-1 are found in CLL cells
from blood [100,101] and lymphoid organs [96]. The data presented
by Dufour et al. [79] indicate that MMP-9-dependent epithelial cell
migration involves the heterodimerization of the PEX domain of
proMMP-9 with CD44, leading to activation of the tyrosine kinase epi-
dermal growth factor receptor (EGFR) and subsequent phosphoryla-
tion of its downstream kinase effectors ERK, AKT and FAK (focal
adhesion kinase) (Fig. 3B). Indeed, EGFR can stimulate various down-
stream cell signaling cascades, including the PI3K/AKT pathway that
favors cell migration and cancer cell invasion [102]. Moreover, FAK
reportedly coordinates cell adhesion, polarization, migration, survival
and death [103]. Chetty and colleagues suggested a role for MMP-2 in
VEGF-induced lung tumor angiogenesis [93]. The interaction of
proMMP-2 with integrin αvβ3 on A549 epithelial cells induces
PI3K/AKT-mediated VEGF expression and related angiogenesis in
vitro (Fig. 3C). The part of MMP-2 that binds to αvβ3 remains to be
determined. Importantly, these results have been validated in vivo
in a spontaneous lung metastasis mouse model [93].
7. Open questions and conclusions
The gelatinases' established functions depend on their proteolytic
activity. By cleaving ECM components, releasing ECM-associated
growth factors, shedding membrane-anchored cytokines and recep-
tors and regulating chemokine activity, gelatinases process signaling
molecules that in turn inuence tumor cell growth, migration, inva-
sion and angiogenesis. The publications detailed in this review high-
light the ability of cell-surface-associated gelatinases to directly
trigger intracellular signaling pathways that control the afore-
mentioned critical cellular processes and behavior. The observed
cell surface association of gelatinases with integrins or other integral
(A) (B) (C)
Fig. 3. Schematic representations of the signaling pathways induced by gelatinases and integral receptors. (A) ProMMP-9 interacts (via its PEX domain) with α4β1 and CD44 on B-
CLL cells, leading to Lyn activation, STAT3 phosphorylation and MCL-1 up-regulation. Mcl-1 (a member of the Bcl-2 family) is essential for lymphocyte survival. (B) The PEX domain
of proMMP-9 interacts with CD44 on tumor epithelial cells (COS-1/kidney, HT1080/brosarcoma, MDA-MB435/breast cancer cells) leading to the activation of EGFR and stimula-
tion of various downstream cell signaling cascades, such as PI3K/AKT, ERK and FAK signals that coordinate cell migration. (C) The interaction of proMMP-2 with integrin αvβ3on
lung cancer cells (A549 cells) activates PI3/AKT signaling, leading to the activation of hypoxia-induced transcription factor-1α(HIF-1α). The latter regulates the expression of the
primarily pro-angiogenic vascular endothelial growth factor VEGF-A. VEGF/VEGFR activation drives vascular sprouting, endothelial cell differentiation and then microtubule
formation.
33B. Bauvois / Biochimica et Biophysica Acta 1825 (2012) 2936
membrane proteins suggests that the signals triggered by these en-
zymes are intertwined with those triggered by integral proteins;
hence, gelatinases may be involved in regulating other aspects of
cell behavior, such as proliferation, differentiation, adhesion and apo-
ptosis. Furthermore, the sheer variety of well-known and newly dis-
covered functions for gelatinases (i.e. secreted forms versus
membrane-bound forms, proforms versus active forms, etc.) begs sev-
eral questions. To what extent are catalytic and non-catalytic activities
(via CBD and PEX) related or interdependent? Are all cell types able to
activate signaling cascades in response to membrane-bound gelati-
nase? Is MMP-2- and MMP-9-mediated outside-in signaling relevant
in other disease states (e.g. inammation and cardiovascular disease)?
Does gelatinase-mediated outside-in signaling extend to other secret-
ed MMPs that might colocalize with integral proteins? At present, it is
thought that proMMP-1 interacts with α2β1 integrin on epithelial
cells [104] and on keratinocytes via its PEX domain [105], whereas
MMP-19 binds to myeloid cells via its PEX domain [106].
Antiproteolytic therapies have sought to target the MMPs' catalyt-
ic activity and thus inhibit tumor progression [25]. The failure of
MMPIs as cancer drugs in the clinic may be explained by their lack
of selectivity towards MMPs (including gelatinases). In light of our
current knowledge of the gelatinases' proteolytic and non-
proteolytic (i.e. outside-in signaling) roles, the enzyme inhibitor ap-
proach may no longer be sufcient because it does not affect the gela-
tinases' interactions with cell surface proteins and consequent
signaling. New therapeutic strategies are focusing on more selective
MMPIs and targeting motifs outside the active site (the exosite)of
individual MMPs; newly designed inhibitors include peptides that
block exosite-mediated cell surface interactions and function-
blocking anti-MMP antibodies [28,107,108]. These approaches have
already been used to target MMP-9 [79,89,109]. A neutralizing anti-
body targeting the PEX domain of MMP-9 bound to LRP-1 at the sur-
face of Schwann cells, blocks cell migration in vitro [109].An
inhibitory peptide that binds selectively to the MMP-9 PEX domain
has already been developed [89]. This small inhibitor prevents PEX
from binding to α1β5 integrin and blocks cell migration in vitro and
tumor xenograft growth in vivo [89]. Synthetic peptides targeting
specic sites of the PEX domain of MMP-9 inhibit the motility of
HT-1080 and MDA-MB-435 tumor cells [79]. These studies indicate
that targeting the PEX domain of MMP-9 by antibodies or peptides
may be a viable approach to abrogate MMP-9-mediated cell function.
In conclusion, recent insights into the potential role of gelatinases as
outside-in signalingmolecules may provide opportunities for the devel-
opment of new gelatinase inhibitors (such as antibodies and peptides)
and validation in relevant animal models, before use as independent
agents or in combination with other cancer treatment strategies.
Acknowledgement
Funding for this work was from the Institut National de la Santé et
de la Recherche Médicale (INSERM).
References
[1] K. Kessenbrock, V. Plaks, Z. Werb, Matrix metalloproteinases: regulators of the
tumor microenvironment, Cell 141 (2010) 5267.
[2] T. Klein, R. Bischoff, Physiology and pathophysiology of matrix metalloproteases,
Amino Acids 41 (2011) 271290.
[3] W. Cruz-Munoz, R. Khokha, The role of tissue inhibitors of metalloproteinases in
tumorigenesis and metastasis, Crit. Rev. Clin. Lab. Sci. 45 (2008) 291338.
[4] D. Bourboulia, W.G. Stetler-Stevenson, Matrix metalloproteinases (MMPs) and
tissue inhibitors of metalloproteinases (TIMPs): positive and negative regulators
in tumor cell adhesion, Semin. Cancer Biol. 20 (2010) 161168.
[5] K. Brew, H. Nagase, The tissue inhibitors of metalloproteinases (TIMPs): an an-
cient family with structural and functional diversity, Biochim. Biophys. Acta
1803 (2010) 5571.
[6] L. Devy, D.T. Dranseld, New strategies for the next generation of matrix-
metalloproteinase inhibitors: selectively targeting membrane-anchored MMPs
with therapeutic antibodies, Biochem. Res. Int. (2011), doi:10.1155/2011/191670.
[7] M. Bjorklund, E. Koivunen, Gelatinase-mediated migration and invasion of can-
cer cells, Biochim. Biophys. Acta 1755 (2005) 3769.
[8] R. Roy, J. Yang, M.A. Moses, Matrix metalloproteinases as novel biomarkers and
potential therapeutic targets in human cancer, J. Clin. Oncol. 27 (2009)
52875297.
[9] K.J. Hateld, H. Reikvam, O. Bruserud, The crosstalk between the matrix metal-
loprotease system and the chemokine network in acute myeloid leukemia,
Curr. Med. Chem. 17 (2010) 44484461.
[10] B. Cauwe, P.E. Van den Steen, G. Opdenakker, The biochemical, biological, and
pathological kaleidoscope of cell surface substrates processed by matrix metal-
loproteinases, Crit. Rev. Biochem. Mol. Biol. 42 (2007) 113185.
[11] D. Rodriguez, C.J. Morrison, C.M. Overall, Matrix metalloproteinases: what do
they not do? New substrates and biological roles identied by murine models
and proteomics, Biochim. Biophys. Acta 1803 (2010) 3954.
[12] E.I. Deryugina, J.P. Quigley, Pleiotropic roles of matrix metalloproteinases in
tumor angiogenesis: contrasting, overlapping and compensatory functions,
Biochim. Biophys. Acta 1803 (2010) 103120.
[13] C. Gialeli, A.D. Theocharis, N.K. Karamanos, Roles of matrix metallo proteinases in
cancer progression and their pharmacological targeting, FEBS J. 278 (2011)
1627.
[14] C.C. Lynch, Matrix metalloproteinases as master regulators of the vicious cycle of
bone metastasis, Bone 48 (2011) 4453.
[15] N.G. Li, Z.H. Shi, Y.P. Tang, J.A. Duan, Selective matrix metalloproteinase inhibi-
tors for cancer, Curr. Med. Chem. 16 (2009) 38053827.
[16] G. Dorman, S. Cseh, I. Hajdu, L. Barna, D. Konya, K. Kupai, L. Kovacs, P.
Ferdinandy, Matrix metalloproteinase inhibitors: a critical appraisal of design
principles and proposed therapeutic utility, Drugs 70 (2010) 949964.
[17] P. Falardeau, P. Champagne, P. Poyet, C. Hariton, E. Dupont, Neovastat, a natural-
ly occurring multifunctional antiangiogenic drug, in phase III clinical trials,
Semin. Oncol. 28 (2001) 620625.
[18] M. Cianfrocca, T.P. Cooley, J.Y. Lee, M.A. Rudek, D.T. Scadden, L. Ratner, J.M.
Pluda, W.D. Figg, S.E. Krown, B.J. Dezube, Matrix metalloproteinase inhibitor
COL-3 in the treatment of AIDS-related Kaposi's sarcoma: a phase I AIDS malig-
nancy consortium study, J. Clin. Oncol. 20 (2002) 153159.
[19] A.N. Sapadin, R. Fleischmajer, Tetracyclines: nonantibiotic properties and their
clinical implications, J. Am. Acad. Dermatol. 54 (2006) 258265.
[20] M.Cianfrocca,S.Lee,J.VonRoenn,M.A.Rudek,B.J.Dezube,S.E.Krown,J.A.
Sparano, Pilot study evaluating the interaction between paclitaxel and prote-
ase inhibitors in patients with human immunodeciency virus-associated
Kaposi's sarcoma: an Eastern Cooperative Oncology Group (ECOG) and AIDS
Malignancy Consortium (AMC) trial, Cancer Chemother. Pharmacol. 68
(2011) 827833.
[21] L. Lacerna, J. Hohneker, Zoledronic acid for the treatment of bone metastases in
patients with breast cancer and other solid tumors, Semin. Oncol. 30 (2003)
150160.
[22] R.K. Yadav, S.P. Gupta, P.K. Sharma, V.M. Patil, Recent advances in studies on
hydroxamates as matrix metalloproteinase inhibitors: a review, Curr. Med.
Chem. 18 (2011) 17041722.
[23] T.G. Mant, D. Bradford, D.M. Amin, J. Pisupati, Y. Kambayashi, Y. Yano, K. Tanaka,
T. Yamada-Sawada, Pharmacokinetics and safety assessments of high-dose and
4-week treatment with S-3304, a novel matrix metalloproteinase inhibitor, in
healthy volunteers, Br. J. Clin. Pharmacol. 63 (2007) 512526.
[24] A.A. Chiappori, S.G. Eckhardt, R. Bukowski, D.M. Sullivan, M. Ikeda, Y. Yano, T.
Yamada-Sawada, Y. Kambayashi, K. Tanaka, M.M. Javle, T. Mekhail, L. O'Bryant,
C.P.J. Creaven, A phase I pharmacokinetic and pharmacodynamic study of
s-3304, a novel matrix metalloproteinase inhibitor, in patients with advanced
and refractory solid tumors, Clin. Cancer Res. 13 (2007) 20912099.
[25] C.M. Overall, O. Kleifeld, Towards third generation matrix metalloproteinase in-
hibitors for cancer therapy, Br. J. Cancer 94 (2006) 941946.
[26] C. Lopez-Otin, L.M. Matrisian, Emerging roles of proteases in tumour suppres-
sion, Nat. Rev. Cancer 7 (2007) 800808.
[27] G. Murphy, H. Nagase, Progress in matrix metalloproteinase research, Mol. As-
pects Med. 29 (2008) 290308.
[28] C.M. Overall, O. Kleifeld, Tumour microenvironment opinion: validating ma-
trix metalloproteinases as drug targets and anti-targets for cancer therapy,
Nat. Rev. Cancer 6 (2006) 227239.
[29] M.D. Martin, L.M. Matrisian, The other side of MMPs: protective roles in tumor
progression, Cancer Metastasis Rev. 26 (2007) 717724.
[30] T. Turpeenniemi-Hujanen, Gelatinases (MMP-2 and -9) and their natural in-
hibitors as prognostic indicators in solid cancers, Biochimie 87 (2005)
287297.
[31] X.F. Yu, Z.C. Han, Matrix metalloproteinases in bone marrow: roles of gelatinases
in physiological hematopoiesis and hematopoietic malignancies, Histol. Histo-
pathol. 21 (2006) 519531.
[32] M. Rydlova, L. Holubec Jr., M. Ludvikova Jr., D. Kalfert, J. Franekova, C. Povysil, M.
Ludvikova, Biological activity and clinical implications of the matrix metallopro-
teinases, Anticancer. Res. 28 (2008) 13891397.
[33] G. Klein, E. Vellenga, M.W. Fraaije, W.A. Kamps, E.S. de Bont, The possible role of
matrix metalloproteinase (MMP)-2 and MMP-9 in cancer, e.g. acute leukemia,
Crit. Rev. Oncol. Hematol. 50 (2004) 87100.
[34] M. Toth, D.C. Gervasi, R. Fridman, Phorbol ester-induced cell surface association
of matrix metalloproteinase-9 in human MCF10A breast epithelial cells, Cancer
Res. 57 (1997) 31593167.
[35] J. Paupert, V. Mansat-De Mas, C. Demur, B. Salles, C. Muller, Cell-surface MMP-9
regulates the invasive capacity of leukemia blast cells with monocytic features,
Cell Cycle 7 (2008) 10471053.
34 B. Bauvois / Biochimica et Biophysica Acta 1825 (2012) 2936
[36] R. Fridman, M. Toth, I. Chvyrkova, S.O. Meroueh, S. Mobashery, Cell surface asso-
ciation of matrix metalloproteinase-9 (gelatinase B), Cancer Metastasis Rev. 22
(2003) 153166.
[37] G.A. Bannikov, T.V. Karelina, I.E. Collier, B.L. Marmer, G.I. Goldberg, Substrate
binding of gelatinase B induces its enzymatic activity in the presence of intact
propeptide, J. Biol. Chem. 277 (2002) 1602216027.
[38] N. Geurts, E. Martens, I. Van Aelst, P. Proost, G. Opdenakker, P.E. Van den Steen,
Beta-hematin interaction with the hemopexin domain of gelatinase B/MMP-9
provokes autocatalytic processing of the propeptide, thereby priming activation
by MMP-3, Biochemistry 47 (2008) 26892699.
[39] C. Freise, U. Erben, M. Muche, R. Farndale, M. Zeitz, R. Somasundaram, M. Ruehl,
The alpha 2 chain of collagen type VI sequesters latent proforms of matrix-
metalloproteinases and modulates their activation and activity, Matrix Biol. 28
(2009) 480489.
[40] J. Oh, R. Takahashi, S. Kondo, A. Mizoguchi, E. Adachi, R.M. Sasahara, S.
Nishimura, Y. Imamura, H. Kitayama, D.B. Alexander, C. Ide, T.P. Horan, T.
Arakawa, H. Yoshida, S. Nishikawa, Y. Itoh, M. Seiki, S. Itohara, C. Takahashi, M.
Noda, The membrane-anchored MMP inhibitor RECK is a key regulator of extra-
cellular matrix integrity and angiogenesis, Cell 107 (2001) 789800.
[41] C. Takahashi, Z. Sheng, T.P. Horan, H. Kitayama, M. Maki, K. Hitomi, Y. Kitaura, S.
Takai, R.M. Sasahara, A. Horimoto, Y. Ikawa, B.J. Ratzkin, T. Arakawa, M. Noda,
Regulation of matrix metalloproteinase-9 and inhibition of tumor invasion by
the membrane-anchored glycoprotein RECK, Proc. Natl. Acad. Sci. U. S. A. 95
(1998) 1322113226.
[42] M. Egeblad, Z. Werb, New functions for the matrix metalloproteinases in cancer
progression, Nat. Rev. Cancer 2 (2002) 161174.
[43] P. Baluk, W.W. Raymond, E. Ator, L.M. Coussens, D.M. McDonald, G.H. Caughey,
Matrix metalloproteinase-2 and -9 expression increases in Mycoplasma-
infected airways but is not required for microvascular remodeling, Am. J. Phy-
siol. Lung Cell Mol. Physiol. 287 (2004) L307L317.
[44] K. Hotary, X.Y. Li, E. Allen, S.L. Stevens, S.J. Weiss, A cancer cell metalloprotease
triad regulates the basement membrane transmigration program, Genes Dev.
20 (2006) 26732686.
[45] F. Sabeh, X.Y. Li, T.L. Saunders, R.G. Rowe, S.J. Weiss, Secreted versus membrane-
anchored collagenases: relative roles in broblast-dependent collagenolysis and
invasion, J. Biol. Chem. 284 (2009) 2300123011.
[46] M. Hangai, N. Kitaya, J. Xu, C.K. Chan, J.J. Kim, Z. Werb, S.J. Ryan, P.C. Brooks, Ma-
trix metalloproteinase-9-dependent exposure of a cryptic migratory control site
in collagen is required before retinal angiogenesis, Am. J. Pathol. 161 (2002)
14291437.
[47] J. Xu, D. Rodriguez, E. Petitclerc, J.J. Kim, M. Hangai, Y.S. Moon, G.E. Davis, P.C.
Brooks, Proteolytic exposure of a cryptic site within collagen type IV is required
for angiogenesis and tumor growth in vivo, J. Cell Biol. 154 (2001) 10691079.
[48] G. Giannelli, J. Falk-Marzillier, O. Schiraldi, W.G. Stetler-Stevenson, V. Quaranta,
Induction of cell migration by matrix metalloprotease-2 cleavage of laminin-5,
Science 277 (1997) 225228.
[49] G. Bergers, R. Brekken, G. McMahon, T.H. Vu, T. Itoh, K. Tamaki, K. Tanzawa, P.
Thorpe, S. Itohara, Z. Werb, D. Hanahan, Matrix metalloproteinase-9 triggers
the angiogenic switch during carcinogenesis, Nat. Cell Biol. 2 (2000) 737744.
[50] Q. Yu, I. Stamenkovic, Cell surface-localized matrix metalloproteinase-9 proteo-
lytically activates TGF-beta and promotes tumor invasion and angiogenesis,
Genes Dev. 14 (2000) 163176.
[51] L. Yang, L.M. DeBusk, K. Fukuda, B. Fingleton, B. Green-Jarvis, Y. Shyr, L.M. Matri-
sian, D.P. Carbone, P.C. Lin, Expansion of myeloid immune suppressor Gr
+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis,
Cancer Cell 6 (2004) 409421.
[52] R. Du, K.V. Lu, C. Petritsch, P. Liu, R. Ganss, E. Passegue, H. Song, S. Vandenberg, R.S.
Johnson, Z. Werb, G. Bergers, HIF1alpha induces the recruitment of bone marrow-
derived vascular modulatory cells to regulate tumor angiogenesis and invasion,
Cancer Cell 13 (2008) 206220.
[53] V.C. Ardi, P.E. Van den Steen, G. Opdenakker, B. Schweighofer, E.I. Deryugina, J.P.
Quigley, Neutrophil MMP-9 proenzyme, unencumbered by TIMP-1, undergoes
efcient activation in vivo and catalytically induces angiogenesis via a basic -
broblast growth factor (FGF-2)/FGFR-2 pathway, J. Biol. Chem. 284 (2009)
2585425866.
[54] Y. Hamano, M. Zeisberg, H. Sugimoto, J.C. Lively, Y. Maeshima, C. Yang, R.O.
Hynes, Z. Werb, A. Sudhakar, R. Kalluri, Physiological levels of tumstatin, a frag-
ment of collagen IV alpha3 chain, are generated by MMP-9 proteolysis and sup-
press angiogenesis via alphaV beta3 integrin, Cancer Cell 3 (2003) 589601.
[55] R. Heljasvaara, P. Nyberg, J. Luostarinen, M. Parikka, P. Heikkila, M. Rehn, T.
Sorsa, T. Salo, T. Pihlajaniemi, Generation of biologically active endostatin frag-
ments from human collagen XVIII by distinct matrix metalloproteases, Exp.
Cell Res. 307 (2005) 292304.
[56] A. Mantovani, P. Allavena, A. Sica, F. Balkwill, Cancer-related inammation, Na-
ture 454 (2008) 436444.
[57] M. Tian, J.R. Neil, W.P. Schiemann, Transforming growth factor-beta and the
hallmarks of cancer, Cell. Signal. 23 (2011) 951962.
[58] E. Levi, R. Fridman, H.Q. Miao, Y.S. Ma, A. Yayon, I. Vlodavsky, Matrix metallopro-
teinase 2 releases active soluble ectodomain of broblast growth factor receptor
1, Proc. Natl. Acad. Sci. U. S. A. 93 (1996) 70697074.
[59] I. Nelissen, E. Martens, P.E. Van den Steen, P. Proost, I. Ronsse, G. Opdenakker,
Gelatinase B/matrix metalloproteinase-9 cleaves interferon-beta and is a target
for immunotherapy, Brain 126 (2003) 13711381.
[60] J.L. Fowlkes, D.M. Serra, H. Nagase, K.M. Thrailkill, MMPs are IGFBP-degrading
proteinases: implications for cell proliferation and tissue growth, Ann. N. Y.
Acad. Sci. 878 (1999) 696699.
[61] T. Takino, N. Koshikawa, H. Miyamori, M. Tanaka, T. Sasaki, Y. Okada, M. Seiki, H.
Sato, Cleavage of metastasis suppressor gene product KiSS-1 protein/metastin
by matrix metalloproteinases, Oncogene 22 (2003) 46174626.
[62] V. Takafuji, M. Forgues, E. Unsworth, P. Goldsmith, X.W. Wang, An osteopontin
fragment is essential for tumor cell invasion in hepatocellular carcinoma, Onco-
gene 26 (2007) 63616371.
[63] B.C. Sheu, S.M. Hsu, H.N. Ho, H.C. Lien, S.C. Huang, R.H. Lin, A novel role of metal-
loproteinase in cancer-mediated immunosuppression, Cancer Res. 61 (2001)
237242.
[64] B. Heissig, K. Hattori, S. Dias, M. Friedrich, B. Ferris, N.R. Hackett, R.G. Crystal, P.
Besmer, D. Lyden, M.A. Moore, Z. Werb, S. Rai, Recruitment of stem and pro-
genitor cells from the bone marrow niche requires MMP-9 mediated release of
kit-ligand, Cell 109 (2002) 625637.
[65] M. Seandel, J. Butler, D. Lyden, S. Rai, A catalytic role for proangiogenic
marrow-derived cells in tumor neovascularization, Cancer Cell 13 (2008)
181183.
[66] E. Fiore, C. Fusco, P. Romero, I. Stamenkovic, Matrix metalloproteinase 9 (MMP-
9/gelatinase B) proteolytically cleaves ICAM-1 and participates in tumor cell re-
sistance to natural killer cell-mediated cytotoxicity, Oncogene 21 (2002)
52135223.
[67] G.A. McQuibban, J.H. Gong, E.M. Tam, C.A. McCulloch, I. Clark-Lewis, C.M. Over-
all, Inammation dampened by gelatinase A cleavage of monocyte chemoattrac-
tant protein-3, Science 289 (2000) 12021206.
[68] P.E. Van den Steen, P. Proost, A. Wuyts, J. Van Damme, G. Opdenakker, Neutro-
phil gelatinase B potentiates interleukin-8 tenfold by aminoterminal processing,
whereas it degrades CTAP-III, PF-4, and GRO-alpha and leaves RANTES and MCP-
2 intact, Blood 96 (2000) 26732681.
[69] U.B. Hofmann, R. Houben, E.B. Brocker, J.C. Becker, Role of matrix metalloprotei-
nases in melanoma cell invasion, Biochimie 87 (2005) 307314.
[70] M. Stefanidakis, E. Koivunen, Cell-surface association between matrix metallo-
proteinases and integrins: role of the complexes in leukocyte migration and
cancer progression, Blood 108 (2006) 14411450.
[71] E. Hahn-Dantona, J.F. Ruiz, P. Bornstein, D.K. Strickland, The low density lipopro-
tein receptor-related protein modulates levels of matrix metalloproteinase 9
(MMP-9) by mediating its cellular catabolism, J. Biol. Chem. 276 (2001)
1549815503.
[72] Z. Yang, D.K. Strickland, P. Bornstein, Extracellular matrix metalloproteinase 2
levels are regulated by the low density lipoprotein-related scavenger receptor
and thrombospondin 2, J. Biol. Chem. 276 (2001) 84038408.
[73] P.E.VandenSteen,I.VanAelst,V.Hvidberg,H.Piccard,P.Fiten,C.Jacobsen,S.K.
Moestrup, S. Fry, L. Royle, M.R. Wormald, R. Wallis, P.M. Rudd, R.A. Dwek, G. Opdenak-
ker, The hemopexin and O-glycosylated domains tune gelatinase B/MMP-9 bioavail-
ability via inhibition and binding to cargo receptors, J. Biol. Chem. 281 (2006)
1862618637.
[74] S. Monferran, J. Paupert, S. Dauvillier, B. Salles, C. Muller, The membrane form of
the DNA repair protein Ku interacts at the cell surface with metalloproteinase 9,
EMBO J. 23 (2004) 37583768.
[75] L.Y. Bourguignon, Z. Gunja-Smith, N. Iida, H.B. Zhu, L.J. Young, W.J. Muller, R.D.
Cardiff, CD44v(3,810) is involved in cytoskeleton-mediated tumor cell migra-
tion and matrix metalloproteinase (MMP-9) association in metastatic breast
cancer cells, J. Cell. Physiol. 176 (1998) 206215.
[76] Q. Yu, I. Stamenkovic, Localization of matrix metalloproteinase 9 to the cell sur-
face provides a mechanism for CD44-mediated tumor invasion, Genes Dev. 13
(1999) 3548.
[77] Y. Hu, L.B. Ivashkiv, Costimulation of chemokine receptor signaling by matrix
metalloproteinase-9 mediates enhanced migration of IFN-alpha dendritic cells,
J. Immunol. 176 (2006) 60226033.
[78] A. Dufour, N.S. Sampson, S. Zucker, J. Cao, Role of the hemopexin domain of ma-
trix metalloproteinases in cell migration, J. Cell. Physiol. 217 (2008) 643651.
[79] A. Dufour, S. Zucker, N.S. Sampson, C. Kuscu, J. Cao, Role of matrix
metalloproteinase-9 dimers in cell migration: design of inhibitory peptides, J.
Biol. Chem. 285 (2010) 3594435956.
[80] J. Redondo-Munoz, E. Ugarte-Berzal, J.A. Garcia-Marco, M.H. del Cerro, P.E.
Van den Steen, G. Opdenakker, M.J. Terol, A. Garcia-Pardo, Alpha4beta1 integ-
rin and 190-kDa CD44v constitute a cell surface docking complex for gelati-
nase B/MMP-9 in chronic leukemic but not in normal B cells, Blood 112
(2008) 169178.
[81] P.C. Brooks, S. Stromblad, L.C. Sanders, T.L. von Schalscha, R.T. Aimes, W.G.
Stetler-Stevenson, J.P. Quigley, D.A. Cheresh, Localization of matrix
metalloproteinase MMP-2 to the surface of invasive cells by interaction with
integrin alpha v beta 3, Cell 85 (1996) 683693.
[82] P.C. Brooks, S. Silletti, T.L. von Schalscha, M. Friedlander, D.A. Cheresh, Disrup-
tion of angiogenesis by PEX, a noncatalytic metalloproteinase fragment with
integrin binding activity, Cell 92 (1998) 391400.
[83] S. Menashi, M. Dehem, I. Souliac, Y. Legrand, R. Fridman, Density-dependent reg-
ulation of cell-surface association of matrix metalloproteinase-2 (MMP-2) in
breast-carcinoma cells, Int. J. Cancer 75 (1998) 259265.
[84] U.B. Hofmann, J.R. Westphal, A.A. Van Kraats, D.J. Ruiter, G.N. Van Muijen, Ex-
pression of integrin alpha(v)beta(3) correlates with activation of membrane-
type matrix metalloproteinase-1 (MT1-MMP) and matrix metalloproteinase-2
(MMP-2) in human melanoma cells in vitro and in vivo, Int. J. Cancer 87
(2000) 1219.
[85] S. Silletti, T. Kessler, J. Goldberg, D.L. Boger, D.A. Cheresh, Disruption of matrix
metalloproteinase 2 binding to integrin alpha vbeta 3 by an organic molecule in-
hibits angiogenesis and tumor growth in vivo, Proc. Natl. Acad. Sci. U. S. A. 98
(2001) 119124.
35B. Bauvois / Biochimica et Biophysica Acta 1825 (2012) 2936
[86] B. Levkau, R.D. Kenagy, A. Karsan, B. Weitkamp, A.W. Clowes, R. Ross, E.W.
Raines, Activation of metalloproteinases and their association with integrins:
an auxiliary apoptotic pathway in human endothelial cells, Cell Death Differ. 9
(2002) 13601367.
[87] M. Rolli, E. Fransvea, J. Pilch, A. Saven, B. Felding-Habermann, Activated integrin
alphavbeta3 cooperates with metalloproteinase MMP-9 in regulating migration
of metastatic breast cancer cells, Proc. Natl. Acad. Sci. U. S. A. 100 (2003)
94829487.
[88] X.Q. Wang, P. Sun, A.S. Paller, Ganglioside GM3 inhibits matrix
metalloproteinase-9 activation and disrupts its association with integrin, J.
Biol. Chem. 278 (2003) 2559125599.
[89] M. Bjorklund, P. Heikkila, E. Koivunen, Peptide inhibition of catalytic and nonca-
talytic activities of matrix metalloproteinase-9 blocks tumor cell migration and
invasion, J. Biol. Chem. 279 (2004) 2958929597.
[90] M. Stefanidakis, M. Bjorklund, E. Ihanus, C.G. Gahmberg, E. Koivunen, Identica-
tion of a negatively charged peptide motif within the catalytic domain of proge-
latinases that mediates binding to leukocyte beta 2 integrins, J. Biol. Chem. 278
(2003) 3467434684.
[91] M. Stefanidakis, T. Ruohtula, N. Borregaard, C.G. Gahmberg, E. Koivunen, Intra-
cellular and cell surface localization of a complex between alphaMbeta2 integrin
and promatrix metalloproteinase-9 progelatinase in neutrophils, J. Immunol.
172 (2004) 70607068.
[92] P.A. Rupp, R.P. Visconti, A. Czirok, D.A. Cheresh, C.D. Little, Matrix metalloprotei-
nase 2-integrin alpha(v)beta3 binding is required for mesenchymal cell invasiv e
activity but not epithelial locomotion: a computational time-lapse study, Mol.
Biol. Cell 19 (2008) 55295540.
[93] C. Chetty, S.S. Lakka, P. Bhoopathi, J.S. Rao, MMP-2 alters VEGF expression via
alphaVbeta3 integrin-mediated PI3K/AKT signaling in A549 lung cancer cells,
Int. J. Cancer 127 (2010) 10811095.
[94] R.E. Nisato, G. Hosseini, C. Sirrenberg, G.S. Butler, T. Crabbe, A.J. Docherty, M.
Wiesner, G. Murphy, C.M. Overall, S.L. Goodman, M.S. Pepper, Dissecting the
role of matrix metalloproteinases (MMP) and integrin alpha(v)beta3 in angio-
genesis in vitro: absence of hemopexin C domain bioactivity, but membrane-
Type 1-MMP and alpha(v)beta3 are critical, Cancer Res. 65 (2005) 93779387.
[95] U.B. Hofmann, J.R. Westphal, E.T. Waas, J.C. Becker, D.J. Ruiter, G.N. van Muijen,
Coexpression of integrin alpha(v)beta3 and matrix metalloproteinase-2
(MMP-2) coincides with MMP-2 activation: correlation with melanoma pro-
gression, J. Invest. Dermatol. 115 (2000) 625632.
[96] J. Redondo-Munoz, E. Ugarte-Berzal, M.J. Terol, P.E. Van den Steen, M. Hernandez
del Cerro, M. Roderfeld, E. Roeb, G. Opdenakker, J.A. Garcia-Marco, A. Garcia-
Pardo, Matrix metalloproteinase-9 promotes chronic lymphocytic leukemia b cell
survival through its hemopexin domain, Cancer Cell 17 (2010) 160172.
[97] X. Hu, P.K. Paik, J. Chen, A. Yarilina, L. Kockeritz, T.T. Lu, J.R. Woodgett, L.B.
Ivashkiv, IFN-gamma suppresses IL-10 production and synergizes with TLR2
by regulating GSK3 and CREB/AP-1 proteins, Immunity 24 (2006) 563574.
[98] J.S. Rao, P. Bhoopathi, C. Chetty, M. Gujrati, S.S. Lakka, MMP-9 short interfering
RNA induced senescence resulting in inhibition of medulloblastoma growth
via p16(INK4a) and mitogen-activated protein kinase pathway, Cancer Res. 67
(2007) 49564964.
[99] P. Bhoopathi, C. Chetty, S. Kunigal, S.K. Vanamala, J.S. Rao, S.S. Lakka, Blockade of
tumor growth due to matrix metalloproteinase-9 inhibition is mediated by se-
quential activation of beta1-integrin, ERK, and NF-kappaB, J. Biol. Chem. 283
(2008) 15451552.
[100] B. Bauvois, J. Dumont, C. Mathiot, J.P. Kolb, Production of matrix
metalloproteinase-9 in early stage B-CLL: suppression by interferons, Leukemia
16 (2002) 791798.
[101] L.S. Chen, K. Balakrishnan, V. Gandhi, Inammation and survival pathways:
chronic lymphocytic leukemia as a model system, Biochem. Pharmacol. 80
(2010) 19361945.
[102] D.L. Wheeler, E.F. Dunn, P.M. Harari, Understanding resistance to EGFR
inhibitors-impact on future treatment strategies, Nat. Rev. Clin. Oncol. 7
(2010) 493507.
[103] M.C. Frame, H. Patel, B. Serrels, D. Lietha, M.J. Eck, The FERM domain: organizing
the structure and function of FAK, Nat. Rev. Mol. Cell Biol. 11 (2010) 802814.
[104] K. Conant, C. St Hillaire, H. Nagase, R. Visse, D. Gary, N. Haughey, C. Anderson,
J. Turchan, A. Nath, Matrix metalloproteinase 1 interacts with neuronal integ-
rins and stimulates dephosphorylation of Akt, J. Biol. Chem. 279 (2004)
80568062.
[105] J.A. Dumin, S.K. Dickeson, T.P. Stricker, M. Bhattacharyya-Pakrasi, J.D. Roby, S.A.
Santoro, W.C. Parks, Pro-collagenase-1 (matrix metalloproteinase-1) binds the
alpha(2)beta(1) integrin upon release from keratinocytes migrating on type I
collagen, J. Biol. Chem. 276 (2001) 2936829374.
[106] S. Mauch, C. Kolb, B. Kolb, T. Sadowski, R. Sedlacek, Matrix metalloproteinase-19
is expressed in myeloid cells in an adhesion-dependent manner and associates
with the cell surface, J. Immunol. 168 (2002) 12441251.
[107] C.M. Overall, C. Lopez-Otin, Strategies for MMP inhibition in cancer: innovations
for the post-trial era, Nat. Rev. Cancer 2 (2002) 657672.
[108] N. Sela-Passwell, G. Rosenblum, T. Shoham, I. Sagi, Structural and functional
bases for allosteric control of MMP activities: can it pave the path for selective
inhibition? Biochim. Biophys. Acta 1803 (2010) 2938.
[109] E. Mantuano, G. Inoue, X. Li, K. Takahashi, A. Gaultier, S.L. Gonias, W.M. Campana,
The hemopexin domain of matrix metalloproteinase-9 activates cell signaling
and promotes migration of Schwann cells by binding to low-density lipoprotein
receptor-related protein, J. Neurosci. 28 (2008) 1157111582.
36 B. Bauvois / Biochimica et Biophysica Acta 1825 (2012) 2936
... They are a family of endopeptidases, of which 24 have been identified in humans [31,32]. Among them are MMP-2 and MMP-9, also known as gelatinases A and B, respectively [33]. The activities of the MMP-9 and MMP-2 genes are related to breast cancer invasion and metastasis, raising the potential to be involved in the disease [34]. ...
Article
Full-text available
Background In women, breast cancer is the second most frequent type of cancer. Looking for new and effective cancer-specific therapies with little to no adverse effects on healthy cells is critical. Objective Minocycline, a second-generation tetracycline, has shown anticancer effects by targeting multiple pathways in various cancers. This study aimed to determine minocycline effects on the cell proliferation, apoptosis, and invasion of the human MCF-7 cells. Methods MTT assay was used to evaluate the cytotoxicity of minocycline on the cells. Flow cytometry was performed to investigate the induction of apoptosis and the cell cycle progression. The expression levels of apoptotic and migration proteins and genes were assessed by western blotting and qRT-PCR. The scratch test was performed to evaluate the anti-migration effect of the drug. Results The results indicated that the IC50 value of minocycline for MCF-7 cells was 36.10 µM. Minocycline treatment caused sub-G1 cell accumulation, indicating a significant apoptotic effect on the MCF-7 cells. Annexin-V/PI staining revealed a significant rise in early and late apoptotic cell percentages. Minocycline up-regulated Bax and Caspase-3 expression and down-regulated Bcl-2 and Pro-Cas3. The scratch test revealed significant anti-migration effects for minocycline. Furthermore, it caused down-regulation of MMP-2 and MMP-9 in a concentration-dependent method. Conclusion These findings further confirmed the anticancer effect of minocycline and highlighted that minocycline maybe considered as potential therapeutic agent for breast cancer treatment.
... MMPs are released as zymogens and activated in various ways, including by other MMPs breaking down the pro-peptide domain [5]. MMPs have proteolytic activity, and therefore, these can aid in invasion and metastasis, as well as alter signaling pathways that regulate biological mechanisms such as cell growth, survival, invasion, inflammation, and angiogenesis [6]. MMPs can catalyze the hydrolysis of a variety of substrates, including cytokine precursors, growth factors, and hormone receptors [7]. ...
Article
The type-II collagenase, also known as matrix metalloproteinase-8 (MMP-8) plays a vital role in a variety of physiological illnesses namely cardiovascular, musculoskeletal, renal, digestive, and respiratory ailments including cancer. However, numerous novel compounds with effective MMP-8 inhibitory activity have been created by research and academic institutions, pharmaceutical companies, and both. Sadly, despite showing promising results, these compounds have a variety of negative side effects. In this context, a number of MMP-8 inhibitors having potential inhibitory activity with different key structural features such as hydroxamates, carboxylic acid, thiol and mercaptosulfide, phosphonate, and pyrimidine-2,4,6-trione have been discussed. Additionally, various ligand-based and structure-based drug designs that have been previously conducted on a variety of MMP-8 inhibitors have been thoroughly reviewed and that can offer important information regarding the development of novel MMP-8 inhibitors in the future. Therefore, the detailed knowledge about the key structural features of the existing MMP-8 inhibitors can be found, and further with the help of drug design strategies, a novel class of collagenase inhibitors may be identified. In light of this, this work may be beneficial for researchers looking into the processes involved in developing and identifying new potential and promising MMP-8 inhibitors.
Article
Full-text available
The role of metalloproteinases (MMPs) in hematological malignancies, like acute myeloid leukemia (AML), myelodysplastic neoplasms (MDS), and multiple myeloma (MM), is well-documented, and these pathologies remain with poor outcomes despite treatment advancements. In this study, we investigated the effects of batimastat (BB-94), an MMP inhibitor (MMPi), in single-administration and daily administration schemes in AML, MDS, and MM cell lines. We used four hematologic neoplasia cell lines: the HL-60 and NB-4 cells as AML models, the F36-P cells as an MDS model, and the H929 cells as a model of MM. We also tested batimastat toxicity in a normal human lymphocyte cell line (IMC cells). BB-94 decreases cell viability and density in a dose-, time-, administration-scheme-, and cell-line-dependent manner, with the AML cells displaying higher responses. The efficacy in inducing apoptosis and cell cycle arrests is dependent on the cell line (higher effects in AML cells), especially with lower daily doses, which may mitigate treatment toxicity. Furthermore, BB-94 activated apoptosis via caspases and ERK1/2 pathways. These findings highlight batimastat’s therapeutic potential in hematological malignancies, with daily dosing emerging as a strategy to minimize adverse effects.
Article
Full-text available
Purpose To determine whether various inflammatory-, angiogenic/anti-angiogenic-, and extracellular matrix remodeling-associated proteins in plasma, alone or in combination with conventional blood-based markers, can predict intra-amniotic inflammation and/or microbial invasion of the amniotic cavity (IAI/MIAC) in women with spontaneous preterm labor (PTL). Methods A total of 193 singleton pregnant women with PTL (23–33 weeks) were included in this retrospective cohort study. Plasma samples were obtained at the time of amniocentesis. Amniotic fluid (AF) was cultured for microorganism detection and consequent MIAC diagnosis. IL-6 levels were determined in AF and used to identify IAI (AF IL-6 ≥ 2.6 ng/mL). Endostatin, haptoglobin, IGFBP-2/3, LBP, M-CSF, MMP-2/8, pentraxin 3, PlGF, S100A8/A9, and VEGFR-1 levels were assayed in plasma samples by ELISA. CRP levels and neutrophil-to-lymphocyte ratio (NLR) were measured. Results Plasma LBP, MMP-8, and S100A8/A9 levels, CRP levels, and NLR were significantly higher, and plasma IGFBP-2 and MMP-2 levels were significantly lower in women with IAI/MIAC than in those without this condition, whereas no baseline variables differed significantly between the two groups. Using a stepwise regression analysis, a noninvasive prediction model for IAI/MIAC was developed, which included plasma LBP, MMP-2, and MMP-8 levels (area under the curve [AUC], 0.785). The AUC for this prediction model was significantly or borderline greater than that of any single factor included in the model. Conclusions IGFBP-2, LBP, MMP-2, MMP-8, and S100A8/A9 may represent valuable plasma biomarkers for predicting IAI/MIAC in women with PTL. Combination of LBP, MMP-2, and MMP-8 expression data can significantly improve the predictive potential for IAI/MIAC.
Article
Full-text available
Kisspeptin (a product of the KISS1 gene and its receptor) plays an important role in obstetrics, gynecology, and cancer cell metastasis and behavior. In hypothalamic-pituitary-gonadal axis and placentation, Kisspeptin/Kisspeptin receptor affects hormone release and represses trophoblast invasion into maternal deciduae. Endometrial cancer is one of the common gynecological cancers and is usually accompanied by metastasis, the risk factor that causes death. Recently, research has demonstrated that Kisspeptin/Kisspeptin receptor expression in aggressive-stage endometrial cancer tissues. However, the detailed mechanism of Kisspeptin/Kisspeptin receptor in regulating the motility of endometrial cancers is not well understood. In this study, we use endometrial cancer cell lines RL95-2, Ishikawa, HEC-1-A, and HEC-1-B as models to explore the molecular mechanism of Kisspeptin on cell motility. First, we discovered that Kisspeptin/Kisspeptin receptor was expressed in endometrial cancer cells, and Kisspeptin significantly regulated the migration and invasion of endometrial cancer cells. Furthermore, we explored the epithelial–mesenchymal transition marker expression and the underlying signals were regulated on Kisspeptin treatment. In conclusion, we suggest that Kisspeptin regulates endometrial cancer cell motility via FAK and Src expression and the ERK1/2, N-Cadherin, E-Cadherin, beta-Catenin, Twist, and matrix metalloproteinase signaling pathways. We expect these molecules could be candidates for the development of new approaches and therapeutic targets.
Article
Full-text available
Using a novel method of N-substituted succinimide ring opening, new N-hydroxybutanamide derivatives were synthesized. These compounds were evaluated for their ability to inhibit matrix metalloproteinases (MMPs) and their cytotoxicity. The iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide showed the inhibition of MMP-2, MMP-9, and MMP-14 with an IC50 of 1–1.5 μM. All the compounds exhibited low toxicity towards carcinoma cell lines HeLa and HepG2. The iodoaniline derivative was also slightly toxic to glioma cell lines A-172 and U-251 MG. Non-cancerous FetMSC and Vero cells were found to be the least sensitive to all the compounds. In vivo studies demonstrated that the iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide had low acute toxicity. In a mouse model of B16 melanoma, this compound showed both antitumor and antimetastatic effects, with a 61.5% inhibition of tumor growth and an 88.6% inhibition of metastasis. Our findings suggest that the iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide has potential as a lead structure for the development of new MMP inhibitors. Our new synthetic approach can be a cost-effective method for the synthesis of inhibitors of metalloenzymes with promising antitumor potential.
Article
Full-text available
Background: Matrix metalloproteinases (MMPs) not only work as enzymes but also as degrading enzymes that have been shown to play an important function in extracellular matrix (ECM) regeneration, including bone regeneration. To generate new bone tissue, bone regeneration or repair relies on a series of regulated processes in which MMPs play an important role. Bone cells express the MMPs in an active state, and these MMPs are assumed to have a crucial role, not only for the viability and functionality of osteoclasts, osteoblasts, and osteocytes but also for the formation and development of chondrocytes. Objective: This study aimed to review and present the roles of matrix metalloproteinases in bone regeneration. Methods: An analysis of the scientific literature on the topics of matrix metalloproteinases in bone regeneration was done on PubMed and Google Scholar. Search results were screened for articles that described or investigated the impacts matrix metalloproteinases have on bones in relation to dentistry. The journals' cited papers were also assessed for relevance and included if they complied with the criteria for inclusion. Accessibility to the full document was one of the prerequisites for admission. Result: Bone regeneration are intricate ongoing processes involving numerous MMPs, especially MMP 2, 9 and 13. MMP-2 appears to alter bone growth through influencing osteoclast and osteoblast activity and proliferation, MMP-9-deficient animals have abnormal bone formation exclusively during endochondral ossification, MMP 13 is responsible for osteoclast receptor activation, has been linked to the breakdown bone resorption. Conclusions: MMP 2, 9, and 13 play a major protective role in osteogenesis and bone regeneration.
Article
Full-text available
During carcinogenesis of pancreatic islets in transgenic mice, an angiogenic switch activates the quiescent vascula- ture. Paradoxically, vascular endothelial growth factor (VEGF) and its receptors are expressed constitutively. Nevertheless, a synthetic inhibitor (SU5416) of VEGF signalling impairs angiogenic switching and tumour growth. Two metalloproteinases, MMP-2/gelatinase-A and MMP-9/gelatinase-B, are upregulated in angiogenic lesions. MMP- 9 can render normal islets angiogenic, releasing VEGF. MMP inhibitors reduce angiogenic switching, and tumour number and growth, as does genetic ablation of MMP-9. Absence of MMP-2 does not impair induction of angiogene- sis, but retards tumour growth, whereas lack of urokinase has no effect. Our results show that MMP-9 is a compo- nent of the angiogenic switch.
Article
Full-text available
The biological roles of the matrix metalloproteinases (MMPs) have been traditionally associated with the degradation and turnover of most of the components of the extracellular matrix (ECM). This functional misconception has been used for years to explain the involvement of the MMP family in developmental processes, cell homeostasis and disease, and led to clinical trials of MMP inhibitors for the treatment of cancer that failed to meet their endpoints and cast a shadow on MMPs as druggable targets. Accumulated evidence from a great variety of post-trial MMP degradomics studies, ranging from transgenic models to recent state-of-the-art proteomics screens, is changing the dogma about MMP functions. MMPs regulate cell behavior through finely tuned and tightly controlled proteolytic processing of a large variety of signaling molecules that can also have beneficial effects in disease resolution. Moreover, net proteolytic activity relies upon direct interactions between the different protease and protease inhibitor families, interconnected in a complex protease web, with MMPs acting as key nodal components. Such complexity renders simple interpretation of Mmp knockout mice very difficult. Indeed, the phenotype of these models reveals the response of a complex system to the loss of one protease rather than necessarily a direct effect of the lack of functional activity of a protease. Such a shift in the MMP functional paradigm, together with the difficulties associated with current methods of studying proteases this highlights the need for new high content degradomics approaches to uncover and annotate MMP activities in vivo and identify novel interactions within the protease web. Integration of these techniques with specifically designed animal models for final validation should lay the foundations for the development of new inhibitors that specifically target disease-related MMPs and/or their upstream effectors that cause deleterious effects in disease, while sparing MMP functions that are protective.
Article
Chemokines are mediators in inflammatory and autoimmune disorders. Aminoterminal truncation of chemokines results in altered specific activities and receptor recognition patterns. Truncated forms of the CXC chemokine interleukin (IL)-8 are more active than full-length IL-8 (1-77), provided the Glu-Leu-Arg (ELR) motif remains intact. Here, a positive feedback loop is demonstrated between gelatinase B, a major secreted matrix metalloproteinase (MMP-9) from neutrophils, and IL-8, the prototype chemokine active on neutrophils. Natural human neutrophil progelatinase B was purified to homogeneity and activated by stromelysin-1. Gelatinase B truncated IL-8(1-77) into IL-8(7-77), resulting in a 10- to 27-fold higher potency in neutrophil activation, as measured by the increase in intracellular Ca++concentration, secretion of gelatinase B, and neutrophil chemotaxis. This potentiation correlated with enhanced binding to neutrophils and increased signaling through CXC chemokine receptor-1 (CXCR1), but it was significantly less pronounced on a CXCR2-expressing cell line. Three other CXC chemokines—connective tissue-activating peptide-III (CTAP-III), platelet factor-4 (PF-4), and GRO-α—were degraded by gelatinase B. In contrast, the CC chemokines RANTES and monocyte chemotactic protein-2 (MCP-2) were not digested by this enzyme. The observation of differing effects of neutrophil gelatinase B on the proteolysis of IL-8 versus other CXC chemokines and on CXC receptor usage by processed IL-8 yielded insights into the relative activities of chemokines. This led to a better understanding of regulator (IL-8) and effector molecules (gelatinase B) of neutrophils and of mechanisms underlying leukocytosis, shock syndromes, and stem cell mobilization by IL-8.
Article
Cellular invasion depends on cooperation between adhesive and proteolytic mechanisms. Evidence is provided that the matrix metalloproteinase MMP-2 can be localized in a proteolytically active form on the surface of invasive cells, based on its ability to bind directly integrin αvβ3. MMP-2 and αvβ3 were specifically colocalized on angiogenic blood vessels and melanoma cells in vivo. Expression of αvβ3 on cultured melanoma cells enabled their binding to MMP-2 in a proteolytically active form, facilitating cell-mediated collagen degradation. In vitro, these proteins formed an SDS-stable complex that depended on the noncatalytic C-terminus of MMP-2, since a truncation mutant lost the ability to bind αvβ3. These findings define a single cell-surface receptor that regulates both matrix degradation and motility, thereby facilitating directed cellular invasion.
Article
In the present study, we have employed a unique breast cancer cell line (Met-1, which was derived from a high metastatic potential tumor in transgenic mice expressing polyomavirus middle T oncogene) to study the role of CD44 variant isoform(s) in the regulation of metastatic breast tumor cell behavior. The results of reverse transcriptase–polymerase chain reaction, Southern blot, nucleotide sequencing, immunoprecipitation, and immunoblot analyses indicated that these cells express a major CD44 isoform (molecular weight ≈260 kDa) containing a v3,8–10 exon insertion (designated as CD44v3,8–10). In addition, we have determined that CD44v3,8–10 binds specifically to the cytoskeletal proteins such as ankyrin. Biochemical analyses, using competition binding assays and a synthetic peptide identical to NGGNGTVEDRKPSEL (a sequence located between aa480 and aa494 of CD44v3,8–10) indicate that this 15-amino acid peptide binds specifically to the cytoskeletal protein ankyrin (but not to fodrin or spectrin). This peptide competes effectively for ankyrin binding to CD44v3,8–10. Therefore, we believe that the sequence 480NGGNGTVEDRKPSE494L, located at the cytoplasmic domain of CD44v3,8–10, is required for the ankyrin binding. We have also detected that CD44v3,8–10-containing Met-1 cells are capable of forming membrane spikes or “invadopodia” structures and undergo active migration processes. Treatments of Met-1 cells with certain agents including anti-CD44v3 antibody, cytochalasin D (a microfilament inhibitor), and W-7 (a calmodulin antagonist), but not colchicine (a microtubule disrupting agent) effectively inhibit “invadopodia” formation and subsequent tumor cell migration. Further analyses using zymography assays and double immunofluorescence staining indicated that CD44v3,8–10 is closely associated with the active form of matrix metalloproteinase, MMP-9, in a complex within “invadopodia” structures. These findings suggest that CD44v3,8–10 plays an important role in linking ankyrin to the membrane-associated actomyosin contractile system required for “invadopodia” formation (coupled with matrix degradation activities) and tumor cell migration during breast cancer progression. J. Cell. Physiol. 176:206–215, 1998. © 1998 Wiley-Liss, Inc.
Article
The zinc-dependent matrix metalloproteinases (MMPs) belong to a large family of structurally homologous enzymes. These enzymes are involved in a wide variety of biological processes ranging from physiological cell proliferation and differentiation to pathological states associated with tumor metastasis, inflammation, tissue degeneration, and cell death. Controlling the enzymatic activity of specific individual MMPs by antagonist molecules is highly desirable, first, for studying their individual roles, and second as potential therapeutic agents. However, blocking the enzymatic activity with synthetic small inhibitors appears to be an extremely difficult task. Thus, this is an unmet need presumably due to the high structural homology between MMP catalytic domains. Recent reports have recognized a potential role for exosite or allosteric protein regions, distinct from the extended catalytic pocket, in mediating MMP activation and substrate hydrolysis. This raises the possibility that MMP enzymatic and non-enzymatic activities may be modified via antagonist molecules targeted to such allosteric sites or to alternative enzyme domains. In this review, we discuss the structural and functional bases for potential allosteric control of MMPs and highlight potential alternative enzyme domains as targets for designing highly selective MMP inhibitors.