Tải bản đầy đủ (.pdf) (14 trang)

Báo cáo khoa học: Localizing matrix metalloproteinase activities in the pericellular environment doc

Bạn đang xem bản rút gọn của tài liệu. Xem và tải ngay bản đầy đủ của tài liệu tại đây (131.24 KB, 14 trang )

MINIREVIEW
Localizing matrix metalloproteinase activities in the
pericellular environment
Gillian Murphy
1
and Hideaki Nagase
2
1 Department of Oncology, University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
2 Department of Matrix Biology, The Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College London, UK
Introduction
Timely alteration of extracellular matrix (ECM) com-
position and the pericellular environment is essential in
many biological processes such as embryonic develop-
ment, morphogenesis, cell migration, differentiation,
apoptosis and tissue remodeling. In diseases such as
cancer [1], atheroma, arthritis, neurodegenerative dis-
eases and various connective tissue diseases, these pro-
cesses become dysregulated. Matrix metalloproteinases
(MMPs) are pivotal effectors of the cellular microenvi-
ronment and modulate cellular activities and tissue
structure throughout development and physiological
and pathological remodeling [2]. All 23 human MMPs
harbor signals that direct them to the endoplasmic
reticulum and hence to the cell surface or to secretion.
The extracellular activities of the MMPs are multiple,
cleaving not only the components of the ECM, but
also many of the bioactive molecules at or around the
cell surface. Because there is considerable overlap in
substrate specificity amongst the MMPs, mechanisms
to preclude redundancy exist. A number of post-secre-
tory regulations of MMP activities have been described


[3]. Specific MMPs do have greater affinity for specific
substrates and the concentrations of the active enzyme
and the preferred substrate relative to other substrates
may be determinants of efficacy [4]. The mechanisms
by which cells can regulate their function in a spatial
fashion pose intriguing questions and are clearly of
critical importance in relation to remodeling events,
Keywords
CD44; collagen; extracellular matrix; integrin;
proteoglycan; receptor; tetraspanin
Correspondence
H. Nagase, Department of Matrix Biology,
The Kennedy Institute of Rheumatology
Division, Faculty of Medicine, Imperial
College London, London W6 8LH UK
Fax: +44 02083834488
Tel: 02083834994
E-mail: ;

(Received 26 June 2010, revised 15
September 2010, accepted 22 September
2010)
doi:10.1111/j.1742-4658.2010.07918.x
Matrix metalloproteinases (MMPs) are a group of structurally related pro-
teolytic enzymes containing a zinc ion in the active site. They are secreted
from cells or bound to the plasma membrane and hydrolyze extracellular
matrix (ECM) and cell surface-bound molecules. They therefore play key
roles in morphogenesis, wound healing, tissue repair and remodeling in dis-
eases such as cancer and arthritis. Although the cell anchored membrane-
type MMPs (MT-MMPs) function pericellularly, the secreted MMPs have

been considered to act within the ECM, away from the cells from which
they are synthesized. However, recent studies have shown that secreted
MMPs bind to specific cell surface receptors, membrane-anchored proteins
or cell-associated ECM molecules and function pericellularly at focussed
locations. This minireview describes examples of cell surface and pericellu-
lar partners of MMPs, as well as how they alter enzyme function and
cellular behaviour.
Abbreviations
ECM, extracellular matrix; GPI, glycosylphosphatidylinositol; HB-EGF, heparin-binding epidermal growth factor; LRP, low-density lipoprotein
receptor-related protein; MMP, matrix metalloproteinase, MT-MMP, membrane-type matrix metalloproteinase; TGF, transforming growth
factor; TIMP, tissue inhibitor of metalloproteinase.
2 FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS
directed cell migration and the directed interactions
with other cells. Among all the MMPs, six are mem-
brane-type (MT)-MMPs that have specific domains
sequestering them at the cell membrane. The majority
of MMPs are secreted into the extracellular space,
although there is accumulating evidence that they may
be recruited back to the local cell environment by
interactions with cell surface proteins and the pericellu-
lar matrix (Table 1). This minireview describes some of
the ingenious devices that have been constructed to
focus and regulate the proteolytic activity of the
MMPs in the pericellular environment.
MMP domain structure
Besides the archetypal secretory signal sequence, regu-
latory propeptide and catalytic domain, the MMPs
have a C-terminal hemopexin-like domain, with the
exception of MMP-7, MMP-23 and MMP-26. MMP-2
and MMP-9 also have three repeats of the fibronectin

type II motif inserted into the catalytic domain. There
are six membrane-anchored MMPs, termed ‘mem-
brane-type’ (MT); four are transmembrane proteins
with short cytoplasmic domains (MT1-, MT2-, MT3-
and MT5-MMP) and two with glycosylphosphatidyli-
nositol (GPI) anchors (MT4- and MT6-MMP) [5].
Although many studies initially focused on the cata-
lytic domain, particularly in relation to the develop-
ment of active site inhibitors, it is now appreciated
that the extracatalytic domains of the MMPs also play
important roles in their function. The N-terminal pro-
peptide acts to maintain the latency of the MMPs by
the presence of a cysteine residue in the active site
coordinated to the catalytic zinc ion [6–9]. The activa-
tion mechanism (known as the ‘cysteine switch’)
involves proteolytic cleavages of the propeptide caus-
ing a destabilization of the cysteine–zinc interaction
[10]. The extra catalytic domains of the MMPs can
contribute to macromolecular substrate specificity; the
fibronectin type II domains of MMP-2 and MMP-9
are important for the cleavage of denatured collagens,
type IV collagen and elastin [11,12]. The hemopexin
Table 1. Cell surface-binding partners of MMPs. Some representative examples of the associations of MMPs with cell surface molecules
and the domain involved in interactions are shown. It is possible that such interactions confer site specificity for MMP action and could form
the basis for therapeutic targeting outside the catalytic cleft.
MMP Domain Cell surface-binding Reference
MMP-1 Linker and Hemopexin a2b1 integrin I-domain [39,101]
MMP-2 Hemopexin MT1-MMP:TIMP-2 complex [102]
MMP-2 Hemopexin Chondroitin 4-sulfate (cell membrane) [48]
MMP-2 Catalytic b2 integrin [103]

MMP-2 FN domain LRP-1 [67]
MMP-2 ? Bone sialoprotein (cell surface) [91]
MMP-3 Hemopexin Collagen I [104]
MMP-7 ? CD44, syndecans, glypicans (heparan sulfate) [61,62,105]
MMP-7 Propeptide CD151 [51]
MMP-7 Catalytic Cholesterol sulfate (cell membrane) [85]
MMP-8 Pro and active forms Neutrophil surface [106]
MMP-9 Hemopexin CD44 [55,72]
MMP-9 Hemopexin Ku dimer (Ku80) [73]
MMP-9 Hemopexin b1 integrin [58]
MMP-9 Catalytic b2 integrin [103]
MMP-9 Hemopexin b5 integrin I-EGF-like domains 2 and 3 [107]
MMP-9 Not known Type VI collagen a2 chain (cell surface) [87]
MMP-9 Linker, hemopexin LRP-1 and -2 [71]
MMP-13 Not known Receptor ⁄ LRP-1 [69]
MMP-14 (MT1-MMP) Hemopexin CD44H stem [25]
MMP-14 Not known ?indirect Claudin [22]
MMP-14 Hemopexin CD63 [53]
MMP-14 Hemopexin MMP-14 [21]
MMP-14 ? CD151 [52]
MMP-15 Hemopexin CD44H [108]
MMP-16 Hemopexin CD44H [108]
MMP-16 Catalytic Chondroitin-4-sulfate (cell surface) [109]
MMP-24 Hemopexin CD44H [108]
MMP-25 Hemopexin CD44H [108]
G. Murphy and H. Nagase MMPs in the pericellular environment
FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS 3
domain of MMP-2 was found to bind chemokines
such as monocyte chemoattractant protein-3 and hence
facilitated its cleavage [13]. The hemopexin domains of

the collagenolytic MMPs (MMP-1, MMP-2, MMP-8,
MMP-13 and MT1-MMP) are essential for cleaving
native triple helical collagen monomers. It has been
shown that collagenases locally unwind the triple helix
to allow access of the individual a chains to the active
site center [14]. Hence, the collagen-binding site may
be composed of elements from both the catalytic
and the hemopexin domains. It is also clear that inter-
domain flexibility is key for the specificity of the colla-
genases and possibly other MMPs [15].
Cell membrane-anchored MMPs
Unlike most of the soluble MMPs, membrane-
anchored MT-MMPs are already active at the cell sur-
face because they are cleaved intracellularly by furin-
like proprotein convertases at their specific recognition
sequence, RX[R ⁄ K]R, located at the C-terminus of the
propeptide [16–18]. MT1-MMP is the best studied of
this subfamily and is known to promote cell invasion
and motility by degrading pericellular ECM molecules
and eliciting the ‘shedding’ of CD44 and syndecan1
ectodomains. It also degrades a plethora of other
extracellular and cell surface proteins [18,19]. Impor-
tantly, MT1-MMP activates proMMP2 via a mecha-
nism in which the tissue inhibitor of MMPs, tissue
inhibitor of metalloproteinase (TIMP)-2, bound to the
MT1-MMP catalytic domain acts as a ‘receptor’ for
proMMP-2 at the cell surface. In this system, modules
within blades III and IV of the hemopexin domain of
proMMP-2 bind to the C-terminal domain of TIMP-2
and this ternary complex formation allows proteolytic

activation of proMMP-2 by an adjacent molecule of
MT1-MMP that is free of TIMP-2. MT1-MMP can
also activate proMMP-13 [20]. It is therefore regarded
as one of the key factors influencing the input of the
cellular microenvironment into cell signaling pathways
[18]. Dimerization of MT1-MMP via the hemopexin
domain is essential for both collagenolysis and effec-
tive proMMP-2 activation and it may be the basis
of its function as an oligomer at the cell surface [21].
Miyamori et al. [22], on the other hand, reported that
claudin-5 at endothelial cell tight junctions recruits
MT1-MMP and proMMP-2 on the cell surface to
achieve elevated focal concentrations, leading to
enhanced proMMP-2 activation independent of
TIMP-2. Similar enhancements of proMMP-2 activa-
tion were reported for other MT-MMPs, suggesting
that clustering is an important factor for proMMP-2
activation [22].
To achieve focal degradation, cells localize MT1-
MMP at lamellipodia, the migration front of the cells
[23–25]. This localization may be achieved by interac-
tion with CD44 and integrins (see below). MMP-2 is
also enriched in invadopodia and it has been suggested
that it could be bound to MT1-MMP through TIMP-2,
as described above.
MT1-MMP is largely sequestered intracellularly in
membrane vesicles and may be derived from newly
synthesized material, or from surface enzyme recycled
through endocytosis. Studies to determine MT1-MMP
localization in invading cells are ongoing, although

they have identified pathways necessary for the forma-
tion of invasive structures and the movement of secre-
tory vesicles involving microtubules and Rab proteins
[26–29]. The short 20 amino acid cytoplasmic domain
of MT1-MMP appears to be of importance in linking
the enzyme proteolytic activity to efficient cell migra-
tion and invasion; several studies have shown that
deletion of this domain markedly reduces cell migra-
tion triggered by the enzyme without affecting the cell
surface proteolytic activity of the enzyme [30–33].
However, there is no universal agreement on this point
because, in some over-expression studies, MT1-MMP
with the cytoplasmic domain deleted can efficiently
drive cell migration through collagen [34]. This may be
a result of overloading of the secretory pathway with
an excess of enzyme, over-riding the normal regulatory
mechanisms. In the same study, the transmembrane
domain was found to be essential because secreted col-
lagenases such as MMP-1 or MT1-MMP without the
transmembrane domain did not drive cell invasion
through a 3D collagen matrix. Membrane-anchoring
of either MMP-1, or even the catalytic domain of
MT1-MMP alone, could drive collagenolytic migra-
tion, albeit with considerably less efficiency than MT1-
MMP itself [34]. Cell invasion through a fibrin gel also
required intact membrane tethered MT1-MMP,
although the tethered catalytic domain alone was not
effective in this case [34]. How the hemopexin domain
interacts with a fibrin substrate has not been studied.
Of the other MT-MMPs, only MT2-MMP has been

shown to play a similar role to MT1-MMP in cell
invasion of collagen I gels [35]. Studies looking at cell
invasion through intact basement membranes have
shown that MT2-MMP and MT3-MMP, as well as
MT1-MMP, may have important roles [36]. These
enzymes can degrade laminin and type IV collagen and
the importance of membrane tethering and the lack
of requirement for the hemopexin domain for cell
invasion were demonstrated. The two GPI-anchored
members of the MT-MMP family, MT4-MMP and
MT6-MMP, have a broad substrate repertoire that is
MMPs in the pericellular environment G. Murphy and H. Nagase
4 FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS
still being defined, including ECM proteins, and have
been shown to activate proMMP-2 and proMMP-9.
MT4-MMP also activates proADAMTS-4 [37]. It is
considered that MT4 and MT6-MMP may have
unique roles, related to their localization in specific cel-
lular microenvironments via the GPI anchor within
lipid rafts of cell membranes [16]. GPI-MT-MMP
activity at the cell surface is also regulated by endocy-
tosis and recycling, as reported for MT1-MMP [16].
There is also some preliminary evidence for homodi-
merization of the GPI-MT-MMPs, although this has
not been studied in depth [16].
Integrins
Studies of wound healing in keratinocyte monolayers
using blocking-antibodies indicated that the proteolytic
activity of MMP-1 is required for migration of human
keratinocytes on native collagen I. Upon wounding,

keratinocytes contact dermal collagen I fibrils and the
interaction with a2b1 integrin stimulates the cell to
produce proMMP-1 [38]. Biochemical and cell-based
studies have further shown that both proMMP-1 and
MMP-1 bind the a2b1 integrin via the I domain of the
a2 integrin subunit and that the linker peptide and the
hemopexin domain of MMP-1 are required for optimal
binding [39]. Basal keratinocytes constitutively express
the a2b1 integrin on their basolateral surfaces and, in
wounds, this receptor accumulates at the forward-basal
tip of migrating keratinocyte in contact with dermal
type I collagen [38,39]. Because the a2b1 integrin binds
native collagen I with high affinity, clustering this inte-
grin at contact points would tightly tether resting
keratinocytes to the dermis, although MMP-1 bound
to a2b1 integrin focally cleaves the collagen matrix.
This results in denaturation of collagen fragments,
which weakens the adhesion to the matrix and allows
keratinocyte migration [38,39]. Hence, a2b1 integrin,
MMP-1 and collagen substrate coordinate together to
drive and regulate migrating keratinocyte during
re-epithelialization. This phenomenon, however, has
not been further studied in vivo, probably because
MMP-13, the predominant collagenase in mice, does
not appear to interact with integrins. It is also not
known whether a similar system operates in other cell
types, although other epithelial cells that move in two
dimensions may utilize MMP-1 in a similar way.
MT1-MMP can be colocalized with b1 integrin in
some cell types [40,41], and this appears to regulate its

function. It is not clear whether this the result of a
direct interaction, although these reports suggest that
MT1-MMP and b1 integrin are functioning in the
same area on the cell surface. In ovarian cancer cells,
antibody-induced clustering of a3b1 integrin stimulates
polarized trafficking and cell surface expression of
MT1-MMP, colocalization to aggregated integrin com-
plexes and activation of pro-MMP-2. In the case of
endothelial cells, MT1-MMP is up-regulated and colo-
calizes with b1 integrin at the intercellular contacts of
confluent cells on b1 integrin-interacting matrices such
as collagen I, fibronectin or fibrinogen. This up-regula-
tion was also shown to be the result of an impairment
of internalization of the cell surface MT1-MMP [42].
On migrating endothelial cells, MT1-MMP was found
to be associated with av b3 integrin at motility-associ-
ated structures and the two proteins could be co-im-
munoprecipitated [42]. It has also been reported that
the integrin avb3 binds MMP-2 via its hemopexin
domain [43,44] and it is colocalized with degraded col-
lagen type I [45]. Physical interaction of MT1-MMP
with avb3 can process the av subunit and increases
outside-in signaling via avb3 [46]. Furthermore, it is
possible that MMP-2 and MT1-MMP activities could
be colocalized through their avb3-binding to modify
integrin-ligand interactions rapidly in situ [42,47]. The
isolated hemopexin domain of MMP-2 was reported to
block angiogenesis in model systems and to inhibit the
interaction of MMP-2 with integrin avb3 [43,44]. How-
ever, there remains some controversy on the direct

binding of av
b3 and the MMP-2 hemopexin domain
[48,49]. Cells expressing avb3 could not use MMP-2
coated surfaces either to attach or spread. It is possible
that specific forms of the integrin and MMP-2 are
involved in binding interactions and that this cannot
always be recapitulated in vitro.
MT1-MMP has been described as interacting with
avb8 to activate transforming growth factor (TGF)-
b1. It has been proposed that, upon ligation of avb8
with latent TGFb (latency associated peptide-b1 ⁄
latency-associated peptide-b1), avb8 and MT1-MMP
become closely associated and form a complex on the
cell surface [50]. The mechanism for this is unknown,
although it is postulated that the interaction may be
indirect because the cell surface appears to be
required for productive interactions and the secreted
forms of avb8 and MT1-MMP did not activate TGF-
b [50]. Because the localization of MT1-MMP in
latency-associated peptide-b1 substrate contacts is
dependent on the presence of b8, it is likely that
avb8–latent TGF b interaction initiates the recruit-
ment of MT1-MMP [50].
Tetraspanins
Tetraspanins are evolutionarily conserved membrane
proteins that tend to associate laterally with one
G. Murphy and H. Nagase MMPs in the pericellular environment
FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS 5
another and to cluster dynamically with numerous
partner proteins in membrane microdomains. Conse-

quently, members of this family are involved in the
coordination of intracellular and intercellular pro-
cesses, including signal transduction, cell proliferation,
adhesion and migration. Recent characterization of tet-
raspanin-enriched microdomains suggests that they
might be specially suited for the regulation of avidity
of adhesion receptors and the compartmentalization of
enzymatic activities. Shiomi et al. [51] reported that
CD151 binds to the propeptide of proMMP-7 and
actives the enzyme on the cell surface. It is considered
to be the result of conformational changes in proM-
MP-7 induced by interacting with CD151, thus
facilitating a spontaneous activation of proMMP-7
pericellularly. The tetraspanin CD151 may also be a
key regulator of MT1-MMP function at the surface of
endothelial cells. MT1-MMP colocalizes with tetra-
spanin CD151 and its associated partner a3b1 integrin
at lateral endothelial cell junctions. Biochemical and
fluorescence resonance energy transfer analyses by
Yan
˜
ez-Mo
´
et al. [52] showed that the MT1-MMP
hemopexin domain associates with CD151 and forms
an a3b1 integrin ⁄ CD151 ⁄ MT1-MMP ternary complex.
Ablation of CD151 expression enhanced MT1-MMP-
mediated activation of MMP-2, although collagen
degradation was reduced around the cell periphery.
MT1-MMP subcellular localization and its inclusion

into detergent-resistant membrane domains, as well as
association with the a3b1 integrin, were affected [52].
Thus, CD151 can finely regulate not only proteolytic
activities of MT1-MMP, but also the sites of action
through complex formation with a3b1.
Another tetraspanin, CD63, a component of late
endosomal and lysosomal membranes, interacts with
MT1-MMP directly through the N-terminus of CD63
and the hemopexin domain [53] and accelerates inter-
nalization and lysosomal degradation of MT1-MMP
[53], thus acting as a further regulator of MT1-MMP
trafficking.
CD44
The hyaluronan receptor CD44, of which some forms
are heavily glycosylated and sulfated, is an important
mediator of cell migration and tissue remodeling
events. CD44v(3,8-10) was reported to be associated
with active MMP-9 within the invadopodia of meta-
static breast cancer cells [54]. The complex links to
ankyrin and the membrane-associated actomyosin con-
tractile system required for ‘invadopodia’ formation,
thus coupling matrix degradation and tumor cell
migration during breast cancer progression [54]. Yu
and Stamenkovic [55] also showed that active MMP-9
can associate with CD44 on mammary carcinoma cells
and activate latent TGFb. TIMP-1 binding to a proM-
MP-9–CD44 complex is also a prerequisite for anti-
apoptotic signaling in erythroid cells [56]. Redondo-
Munoz et al. [57] found that CD44v and a4b1integrin
colocalize with MMP-9 in invading lymphoid cells,

and MMP-9 produced by chronic lymphocytic leuke-
mia B cells is considered to contribute to their tissue
infiltration by degrading extracellular and membrane-
anchored substrates. This interaction is mediated by
the hemopexin domain. Binding of soluble or immobi-
lized MMP-9, or the MMP-9 hemopexin domain, to
a4b1 integrin and CD44v prevents B-cell leukemia
lymphocyte apoptosis by inducing Lyn activation,
STAT3 phosphorylation and Mcl-1 up-regulation [58],
although the target(s) of MMP-9 activity in this con-
text are not yet known.
As discussed above, CD44 also interacts with MT1-
MMP, and directs the presence of MT1-MMP in
lamellipodia of the invasive front of migrating cells
[25]. This localization may be achieved by interaction
with CD44 through the hemopexin domain of the
enzyme and stem region of CD44, and CD44 associ-
ates with F-actin through its cytoplasmic domain by
interacting with Ezrin ⁄ Radixin ⁄ Moesin proteins. MT1-
MMP is also localized to F-actin-rich invasive struc-
tures found in some cells, termed invadopodia, and
detailed time-lapse studies have demonstrated that
cortactin and actin aggregates at membrane regions
adherent to matrix where MT1-MMP accumulates
[26]. Matrix degradation leads to cortactin dissociation
from the area, although MT1-MMP remains
associated with foci of degraded matrix [26]. Hence,
proteolytic shedding of CD44 from the cell surface by
MT1-MMP promotes cell migration on a hyaluronan
based 2D matrix [59]. MT1-MMP may therefore act to

regulate the adhesion properties of cellular lamellipo-
dia. Again, the hemopexin domain of MT1-MMP is
required for CD44 shedding and cell migration to
occur in this model. Marrero-Diaz et al. [60] carried
out time-lapse confocal microscopy and fluorescence
resonance energy transfer imaging of carcinoma cells
embedded in a 3D collagen I matrix containing hyal-
uronan and showed that MT1-MMP interacted with
CD44 preferentially at the trailing edge of the invading
tumor cells and that the proteolytic processing of the
CD44 extracellular domain was enriched in the
retracting rear ends. It was concluded that the role of
MT1-MMP in CD44-mediated tumor-cell invasion is
cell retraction, although CD44 is not essential for MT1-
MMP-mediated invasion into the 3D matrix of hyaluro-
nan-collagen.
MMPs in the pericellular environment G. Murphy and H. Nagase
6 FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS
Membrane proteoglycans and
glycosaminoglycans
Proteoglycans, which contain either heparan sulfate or
chondroitin sulfate glycosaminoglycan chains attached
to a protein core, are an important class of cell surface
and ECM molecules regulating activation and activity
of MMPs. Many MMPs are bound to tissues through
interaction with glycosaminoglycans, and MMP-7 is
one of the most tightly bound. MMP-7 is often found
to be bound to heparan sulfate proteoglycans on or
around epithelial cells and in the underlying basement
membrane, and it may be released by heparinase diges-

tion [61]. When it is bound to heparin, the activity of
MMP-7 is greatly enhanced. Two putative heparin-
binding peptides were identified near the C- and N-ter-
minal regions of proMMP-7, although molecular mod-
eling suggested an extensive binding track crossing
multiple peptide strands. Evidence was also found for
the binding of MMP-2, -9, -13 and -16 [48,61]. As sug-
gested by Ra and Parks [4], binding of those MMPs to
heparan sulfate in the extracellular space could prevent
the loss of secreted enzyme, provide a reservoir of
latent enzyme, and facilitate cellular sensing and regu-
lation of enzyme levels. Furthermore, binding to the
cell surface could position the enzyme for directed pro-
teolytic attack for activation of (or by) other MMPs
and for regulation of other cell surface proteins.
Forms of the hyaluronan receptor CD44 bearing
sulfated glycosaminoglycans bind MMP-9, as discussed
above. Yu et al. [62] reported that such forms of
CD44 recruit proteolytically active MMP-7 and the
substrate heparin-binding epidermal growth factor pre-
cursor (proHB-EGF) via the sulfated sugars, forming
a complex on the surface of tumor cell line. The pro-
HB-EGF within this complex is processed by MMP-7,
and the resulting mature HB-EGF engages and acti-
vates its receptor, ErbB4, leading to cell survival. In
CD44() ⁄ )) mice, postpartum uterine involution is
accelerated and maintenance of lactation is impaired
as a result of altered MMP-7 localization and
decreased ErbB4 activation in both uterine and mam-
mary epithelia [62]. Because MMP-7 is known as an

important regulator of many proteolytic events at the
cell surface, it is possible that CD44 interaction could
be a means of localizing the enzyme to key sites. It is
likely that charge interactions with sulfated glycosami-
noglycans of CD44 are important, although the mole-
cular nature of this is not fully understood.
Syndecans and glypicans are other examples of
membrane associated proteoglycans with highly sul-
fated glycosaminoglycan chains that regulate cell sur-
face events by interactions with effectors, including
growth factors, integrins and proteinase inhibitors.
Endometrial epithelial cells and carcinoma cells from
various tissues bind to active MMP-7 at the cell sur-
face. MMP-7-binding could be decreased by interfering
with heparan sulfate proteoglycans, and by interacting
with TIMP-2 or a synthetic MMP inhibitor. The
bound MMP-7 remains fully active towards a macro-
molecular substrate but is resistant to inhibition by
TIMP-2 [63]. MMP-9 associated with the heparan sul-
fate chains of the GPI-anchored cell surface proteogly-
can glypican of murine colon adenocarcinoma cells
[64]. This allows the accumulation of MMP-9 on the
tips of invasive protrusions of the cells and promotes
their motility. Treatment of the cells with heparitinase-I
or heparin released MMP-9 from the cell surface,
which resulted in the suppression of their motility to a
level similar to that exhibited by an MMP inhibitor.
However, the heparan sulfate-interacting domain of
MMP-9 is not known. Iida et al. [48] reported that
melonoma cell specific cell surface chondrotin sulfate

proteoglycan enhances the activation of proMMP-2 by
MT3-MMP and cell invasion in vitro. The complex is
formed through glycosaminoglycan components inter-
acting with the catalytic domain of MT3-MMP and
the hemopexin domain of proMMP-2. This effect was
also observed with isolated chondroitin 4-sulfate, but
not chondroitin 6-sulfate, hyaluronan or heparin, sug-
gesting that a specific sulfation pattern is important in
those reactions.
Low-density lipoprotein
receptor-related protein (LRP) and Ku
LRP is a member of the low-density lipoprotein recep-
tor superfamily and a heterodimeric endocytic receptor
for a large number of proteins, and also has signaling
properties. LRP internalizes many diverse ligands,
including a
2
-macroglobulin-proteinase complexes, sev-
eral serine proteinases, proteinase inhibitors and pro-
teinase–inhibitor complexes [65]. MMP-2, MMP-9 and
MMP-13 have been reported to be endocytosed by
LRP, introducing another level of regulation of peri-
cellular proteolysis by MMPs. ProMMP-2 by itself has
a relatively low affinity to LRP but forms complexes
with either thrombospondin [66] or TIMP-2 [67]
that are readily endocytosed by LRP. The study of
proMMP-2–TIMP-2 complex internalization indicated
that both the fibronectin II domain of MMP-2 and
TIMP-2 have independent binding sites in LRP, which
enhances the uptake of the complex [67]. ProMMP-9 is

internalized as the proMMP-9–TIMP-1 complex [68].
Internalization of MMP-13 is initiated by binding to
an unidentified 170 kDa receptor and the enyzme is
G. Murphy and H. Nagase MMPs in the pericellular environment
FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS 7
transferred to LRP for internalization and intracellular
degradation [69]. Troeberg et al. [70] reported that
TIMP-3, is also internalized by LRP, which also
requires a sulfated proteoglycan as a co-receptor.
The hemopexin domain of MMP-9 contains a bind-
ing site for LRP-1 and LRP-2 and these receptors have
been implicated in regulating MMP-9 activity [71]. The
64 amino acid linker region of MMP-9 between the
catalytic and hemopexin domains is heavily O-glycosy-
lated. The linker region is required to correctly orient
the hemopexin domain for inhibition by TIMP-1 and
internalization by LRP-1 and LRP-2, hence regulating
active enzyme bioavailability [71,72].
Another interesting molecule interacting with MMP-9
on the cell surface is the heterodimeric DNA repair
molecule Ku (Ku70 ⁄ Ku80). Ku not only is present in
the cytosol and nucleus, but also is found on cell
surface of certain cell types. Monferran et al. [73]
reported that MMP-9 is bound to Ku on the leading
and tailing edge of the leukemic cell surface and helps
the cell to migrate into type IV collagen matrices, indi-
cating the importance of the membrane bound Ku in
ECM turnover. The interaction of MMP-9 and Ku is
mediated by the hemopexin domain of MMP-9 and
Ku80.

Emmprin
Emmprin, CD147 (Basigin) is an important cell surface
bound MMP regulator. It is a transmembrane glyco-
protein with two Ig-like domains [74]. It was identified
as an MMP-inducer expressed on epithelial cells and is
known to enhance cell proliferation and multidrug
resistance by promoting hyaluronan synthesis and
angiogenesis via the augmentation of vascular endothe-
lial growth factor production [75]. It interacts with sev-
eral molecules including caveolin, cyclophilin 60 and
monocarboxylate transporters [75]. Although epithelial
Emmprin stimulates surrounding stromal cells to pro-
duce a number of proMMPs, proMMP-1 was found
to interact with Emmprin on human lung carcinoma
cells [76], and both proMMP-1 and active MMP-1
bind to Emmprin on glandular epithelium in the
human endometrium [77] Although the activity of
MMP-1 on the cell surface has not been examined, it
may be another way to specifically regulate pericellular
collagenolytic activity.
Endo180
Endo180 was originally identified as constitutively
recycling cell surface receptor [78]. It was also found
as a macrophage mannose receptor type C lectin [79]
and as urokinase-type plasminogen activator receptor
associated protein [80]. It has also been characterized
as a collagen-binding and collagen-internalization
receptor [81]. MT1-MMP was shown to have a critical
role in collagen phagocytosis [82] and recent studies by
Messaritou et al. [83] have demonstrated that Endo180

is a negative regulator of MT1-MMP activity and thus
down-regulates MT1-MMP-dependent MMP-2 activa-
tion in HT1080 cells. Depletion of Endo180 by siRNA
led to the accumulation of collagen in the medium as a
result of reduced collagen endocytosis, and resulted in
the collagen-dependent increase of MT1-MMP activity
on the cell surface. However, Messaritou et al. [83]
could not show direct binding of Endo180 and MT1-
MMP, suggesting that the effect of Endo180 on the
regulation of MT1-MMP activity involves another
molecule. Their study indicates an intricate coordina-
tion of collagen clearance in the pericellular environ-
ment mediated both collagen internalization and
regulated MT1-MMP activity.
Cholesterol sulfate
MMP-7 cleaves many ECM molecules and other pro-
teins in the cellular microenvironment and is consid-
ered to have ‘sheddase’ activities comparable with
those of the disintegrin MMPs. It is known to induce
adhesion of colon cancer cells by the cleavage of cell
surface proteins, although binding of MMP-7 to cell
surface cholesterol sulfate is essential for this activity
and the induction of cell aggregation [84]. The choles-
terol sulfate-binding site has been identified on the
opposite side of the catalytic cleft of MMP-7 [85], and
it has been proposed that this makes it possible for the
enzyme to cleave both cell surface protein substrates
and those in the pericellular ECM.
Pericellular ECM
MMP-binding to the extracellular matrix has been

noted in many immunohistochemical and biochemical
extraction studies and examples of the potential effects
of matrix macromolecule association with MMPs on
the activity of the latter have been postulated Because
many ECM proteins are associated with the cell sur-
face, it is likely that these act as important binding
sites and modulators of pericellular proteolysis. The
gelatinases, MMP-2 and MMP-9, were the first MMPs
to be recognized as binding to fibrillar collagens such
as type I and their denatured forms within the ECM.
It was shown that this was effected by a ‘gelatin-bind-
ing domain’, consisting of three repeats of the fibro-
nectin type II motif inserted into the catalytic domain
MMPs in the pericellular environment G. Murphy and H. Nagase
8 FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS
[11,86]. Subsequently, proMMP-9 and its TIMP-1
complex were shown to bind with high affinity to a
number of cell lines via cell surface a2 chain of type
IV collagen [87]. Binding of pro-MMP-9 to cells does
not result in zymogen activation and is not followed
by ligand internalization, even after complex formation
with TIMP-1. Interestingly, the proenzyme does not
bind to secreted triple-helical collagen IV. It was pro-
posed that this unique interaction between pro-MMP-9
and a2(IV) may play a role in targeting the zymogen
to cell matrix contacts and in the degradation of the
collagen IV network. Preliminary studies have impli-
cated the gelatin-binding domain of MMP-9 in a2(IV)
collagen-binding [88]. However, the nature of a2(IV)
collagen association to cells is itself not clear. Owen

et al. [89] showed that MMP-9 secreted by activated
PMN leucocytes binds to the cell surface by an
unknown mechanism. Significantly, the bound enzyme
is fully functional proteolytically, although is no longer
regulated by TIMP-1 or TIMP-2. Bannikov et al. [90]
found that the gelatinolytic activity of MMP-9 could
be detected in situ in tissue sections of term placenta,
However, all the enzyme extracted from this tissue was
in a proform. They found that purified proMMP-9
acquired activity against gelatin substrates, although
its propeptide remained intact. These results suggest
that, although activation of all known MMPs in vitro
is accomplished by proteolytic processing of the pro-
peptide, other mechanisms, such as binding to a ligand
or to a substrate, may lead to a disengagement of the
propeptide from the active center of the enzyme, caus-
ing its activation. This observation could have implica-
tions for the association of other MMPs to cell surface
molecules. Bone sialoprotein is a member of a family
of glycoprotein ligands for integrins and can therefore
be cell surface associated. Bone sialoprotein has
been shown to induce limited gelatinase activity in
proMMP-2 without removal of the propeptide and to
restore enzymatic activity to MMP-2 previously inhib-
ited by TIMP-2 [91].
More recently Freise et al. [87] found that proforms
of MMPs were closely associated with collagenous sep-
tae in fibrotic liver tissue and that the triple helical
domain of a2 chain of collagen VI bound with nanom-
olar affinity to procollagenases (proMMP-1, -8 and

-13) and proMMP-3, as well as proMMP-2 and -9.
The binding of collagen VI to those zymogens or acti-
vated MMPs reduced the levels of auto-activation and
enzymatic activities, respectively, with the exception of
an observed increase in proMMP-2 activation and
MMP-2 activity. It was suggested that the a2(VI) chain
modulates MMP availability by sequestering proMMPs
in the ECM and blocking proteolytic activity. Using
tandem affinity expression tagged MMP-13 hinge-
hemopexin domains as a bait, Zhang et al. [92] found
that they bound TIMP-1 and a2-macroglobulin, fibro-
nectin, type VI collagen, xylosyltransferase 1, decorin,
syndecan 4 and serglycin in the medium of human
chondrocytes in culture. Although the consequences of
these interactions remain to be demonstrated, studies
suggest that MMP-13 activity may be either targeted
at a more specific site in the connective tissue matrix,
or that matrix proteins may regulate its activity during
cartilage degradation.
The mechanisms of interaction of the collagenases
with fibrillar collagen substrates is key to directed
cleavage. As exemplified with MT1-MMP, cell surface
collagenolysis is implicated in directional cell move-
ment along the collagen fibrils. In the tissue, interstitial
collagen forms insoluble fibrils that are highly resistant
to digestion, even by collagenases such as MMP-1,
MMP-13 and MT1-MMP. This may be particularly a
result of collagenase cleavage sites in collagen fibrils
being covered mainly by the C-terminal telopeptide of
the neighboring collagen molecule, as shown in the 3D

structure of rat tendon collagen fibrils recently solved
using X-ray diffraction analysis [93,94]. This suggests
that the removal of the C-telopeptide or its structural
alteration needs to take place before collagenase can
act on collagen fibrils. This would also remove impor-
tant cross linking sites from the fibrils. On the other
hand, Saffarian et al. [95] reported that active MMP-1
moves to one direction on collagen fibrils analgous to a
molecular ratchet, which is driven by proteolysis. This
directional movement may be explained by the Orgel
model of collagen fibrils [93]: upon destabilization or
cleavage of the C-telopeptide, collagenase can remove
the C-terminal ¼ fragment including the C-telopep-
tide, which then exposes the C-terminally adjacent col-
lagenase-cleavage site. Subsequent cleavage of this site
will expose another site on the C-terminal side. Such
directional movement of the collagenase may promote
cells to move in one direction when the enzyme is
attached to the cell surface. This may also occur with
MT-MMPs and MMP-1 attached to Emmprin or a2b1
integrin. Cells such as inflammatory cells may use such
a mechanism to move along one direction depending
on the orientation of collagen fibrils. In Orgel’s model,
the a2b1 integrin-binding site in collagen I is also
blocked by the adjacent collagen molecule [96]. To
make these sites available to the integrins, collagenoly-
sis and denaturation of the
¾
fragment is probably
necessary. Further investigations are necessary to

determine how pericellular collagenolysis and integrin
engagement are coordinated during the cell movement
on collagen fibrils. Those questions may apply to the
G. Murphy and H. Nagase MMPs in the pericellular environment
FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS 9
migration of many cell types, including cancer cells,
inflammatory cells, smooth muscle cells and endothe-
lial cells, as well as the directionality of collagen fibres
in the tissue.
Conclusions and future prospects
Subsequent to the discovery of vertebrate collagenase
in the tadpole tail undergoing metamorphosis [97],
MMPs have largely been characterized in relation to
their respective abilities to degrade ECM molecules.
However, for the last two decades, researchers have
recognized that non-ECM proteins, such as serpins,
cytokines, chemokines, growth factors and growth fac-
tor-binding proteins, are also MMP substrates [2].
Along with the cryptic function of ECM molecules
exposed by MMP cleavage, these observations empha-
size that MMPs play diverse roles in many cellular
activities, such as proliferation, differentiation, migra-
tion and apoptosis. The proteolytic actions associated
with these events most effectively occur at or close to
the cell surface. Thus, the MT-MMPs have been
regarded as prime candidates in such activities [19].
However, numerous studies have demonstrated that
secreted MMPs are also recruited to the cell surface,
interacting with cell surface receptors or pericellular
macromolecules, including those of the ECM (Table 1).

Such interactions introduce intricate regulatory mecha-
nisms that create gradients or directionality of prote-
olytic activity. Of particular note is the role of
interacting proteins in endocytic mechanisms to down-
regulate MMP function. Although only a few MMPs
have been so far studied, the diversity of mechanisms
is proving to be enormous. These examples have led us
to consider that many, if not all, of the MMPs may
frequently function pericellularly, rather than at sites
distal to the cells, where the communication between
the cells and the surrounding ECM and other bioactive
molecules takes place. For example, MMP-19 is
another member found to be associated with myeloid
cell surface, although the binding partner is yet to be
identified [98]. Identification of the partner molecule
would help us understand the role of MMP-19 in
blood-derived cell migration. Careful immunocyto-
chemical studies of more recently discovered MMPs
certainly deserve further attention, which may help to
elucidate the molecular mechanism of the proposed
function or introduce new biological functions. The
use of modern techniques for protein identification will
also further the characterization of proteins that inter-
act with MMPs [99,100] and further define cell surface
focussing and regulation of their function in relation
to specific cellular activities.
Acknowledgements
We would like to thank all the individuals who have
contributed to MMP research and apologize that we
have been unable to cite all the relevant studies in this

minireview. Our work is supported by grants from
Cancer Research UK, Medical Research Council,
European Union Framework 6 Program to G.M., the
Wellcome Trust, Arthritis Research UK and National
Institutes of Health (AR40994) to H.N.
References
1 Gialeli C, Theocharis AD & Karamanos NK (2010)
Metalloproteinases in health and disease: roles of
matrix metalloproteinases in cancer progression and
their pharmacological targeting. FEBS J 278, 16–27.
2 Murphy G & Nagase H (2008) Progress in matrix
metalloproteinase research. Mol Aspects Med 29, 290–
308.
3 Hadler- Olsen EFB, Sylte I, Uhlin-Hansen L &
Winberg J-O (2010) Metalloproteinases in health and
disease: regulation of matrix metalloproteinase activity.
FEBS J 278, 28–45.
4 Ra HJ & Parks WC (2007) Control of matrix
metalloproteinase catalytic activity. Matrix Biol 26,
587–596.
5 Nagase H, Visse R & Murphy G (2006) Structure and
function of matrix metalloproteinases and TIMPs.
Cardiovasc Res 69, 562–573.
6 Becker JW, Marcy AI, Rokosz LL, Axel MG,
Burbaum JJ, Fitzgerald PM, Cameron PM, Esser CK,
Hagmann WK, Hermes JD et al. (1995) Stromelysin-1:
three-dimensional structure of the inhibited catalytic
domain and of the C-truncated proenzyme. Protein Sci
4, 1966–1976.
7 Morgunova E, Tuuttila A, Bergmann U, Isupov M,

Lindqvist Y, Schneider G & Tryggvason K (1999)
Structure of human pro-matrix metalloproteinase-2:
activation mechanism revealed. Science 284, 1667–
1670.
8 Elkins PA, Ho YS, Smith WW, Janson CA,
D’Alessio KJ, McQueney MS, Cummings MD &
Romanic AM (2002) Structure of the C-terminally
truncated human ProMMP9, a gelatin-binding matrix
metalloproteinase. Acta Crystallogr D Biol Crystallogr
58, 1182–1192.
9 Jozic D, Bourenkov G, Lim NH, Visse R, Nagase H,
Bode W & Maskos K (2005) X-ray structure of human
proMMP-1: new insights into procollagenase
activation and collagen binding. J Biol Chem 280,
9578–9585.
10 Van Wart HE & Birkedal-Hansen H (1990) The cyste-
ine switch: a principle of regulation of metalloprotein-
ase activity with potential applicability to the entire
MMPs in the pericellular environment G. Murphy and H. Nagase
10 FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS
matrix metalloproteinase gene family. Proc Natl Acad
Sci USA 87, 5578–5582.
11 Murphy G, Nguyen Q, Cockett MI, Atkinson SJ,
Allan JA, Knight CG, Willenbrock F & Docherty AJP
(1994) Assessment of the role of the fibronectin-like
domain of gelatinase A by analysis of a deletion
mutant. J Biol Chem 269, 6632–6636.
12 Shipley JM, Doyle GAR, Fliszar CJ, Ye QZ, Johnson
LL, Shapiro SD, Welgus HG & Senior RM (1996)
The structural basis for the elastolytic activity of the

92-kDa and 72-kDa gelatinases. J Biol Chem 271,
4335–4341.
13 McQuibban GA, Gong JH, Tam EM, McCulloch CA,
Clark-Lewis I & Overall CM (2000) Inflammation
dampened by gelatinase A cleavage of monocyte
chemoattractant protein-3. Science 289, 1202–1206.
14 Chung L, Dinakarpandian D, Yoshida N, Lauer-
Fields JL, Fields GB, Visse R & Nagase H (2004)
Collagenase unwinds triple-helical collagen prior to
peptide bond hydrolysis. EMBO J 23, 3020–3030.
15 Sela-Passwell N, Rosenblum G, Shoham T & Sagi I
(2010) Structural and functional bases for allosteric
control of MMP activities: can it pave the path for
selective inhibition? Biochim Biophys Acta 1803, 29–38.
16 Sohail A, Sun Q, Zhao H, Bernardo MM, Cho JA &
Fridman R (2008) MT4-(MMP17) and MT6-MMP
(MMP25), A unique set of membrane-anchored matrix
metalloproteinases: properties and expression in can-
cer. Cancer Metastasis Rev 27, 289–302.
17 Zucker S, Pei D, Cao J & Lo
´
pez-Otı
´
n C (2003) Mem-
brane type-matrix metalloproteinases (MT-MMP).
Curr Top Dev Biol 54, 1–74.
18 Itoh Y & Seiki M (2006) MT1-MMP: a potent modi-
fier of pericellular microenvironment. J Cell Physiol
206, 1–8.
19 Barbolina MV & Stack MS (2008) Membrane type 1-

matrix metalloproteinase: substrate diversity in pericel-
lular proteolysis. Semin Cell Dev Biol 19, 24–33.
20 Kna
¨
uper V, Bailey L, Worley JR, Soloway P,
Patterson ML & Murphy G (2002) Cellular activation
of proMMP-13 by MT1-MMP depends on the
C-terminal domain of MMP-13. FEBS Lett 532, 127–
130.
21 Itoh Y, Ito N, Nagase H, Evans RD, Bird SA & Seiki
M (2006) Cell surface collagenolysis requires homodi-
merization of the membrane-bound collagenase MT1-
MMP. Mol Biol Cell 17, 5390–5399.
22 Miyamori H, Takino T, Kobayashi Y, Tokai H, Itoh
Y, Seiki M & Sato H (2001) Claudin promotes activa-
tion of pro-matrix metalloproteinase-2 mediated by
membrane-type matrix metalloproteinases. J Biol
Chem 276, 28204–28211.
23 Sato H, Okada Y & Seiki M (1997) Membrane-type
matrix metalloproteinases (MT-MMPs) in cell
invasion. Thromb Haemost 78, 497–500.
24 Itoh Y, Takamura A, Ito N, Maru Y, Sato H,
Suenaga N, Aoki T & Seiki M (2001) Homophilic
complex formation of MT1-MMP facilitates
proMMP-2 activation on the cell surface and promotes
tumor cell invasion. EMBO J 20, 4782–4793.
25 Mori H, Tomari T, Koshikawa N, Kajita M, Itoh Y,
Sato H, Tojo H, Yana I & Seiki M (2002) CD44
directs membrane-type 1 matrix metalloproteinase to
lamellipodia by associating with its hemopexin-like

domain. EMBO J 21, 3949–3959.
26 Artym VV, Zhang Y, Seillier-Moiseiwitsch F,
Yamada KM & Mueller SC (2006) Dynamic interac-
tions of cortactin and membrane type 1 matrix
metalloproteinase at invadopodia: defining the stages
of invadopodia formation and function. Cancer Res
66, 3034–3043.
27 Poincloux R, Lizarraga F & Chavrier P (2009) Matrix
invasion by tumour cells: a focus on MT1-MMP
trafficking to invadopodia. J Cell Sci
122, 3015–3024.
28 Clark ES & Weaver AM (2008). A new role for
cortactin in invadopodia: regulation of protease
secretion. Eur J Cell Biol.
29 Bravo-Cordero JJ, Marrero-Diaz R, Megı
´
as D, Genı
´
s
L, Garcı
´
a-Grande A, Garcı
´
a MA, Arroyo AG &
Montoya MC (2007) MT1-MMP proinvasive activity
is regulated by a novel Rab8-dependent exocytic
pathway. EMBO J 26, 1499–1510.
30 Gingras D, Bousquet-Gagnon N, Langlois S,
Lachambre MP, Annabi B & Be
´

liveau R (2001)
Activation of the extracellular signal-regulated protein
kinase (ERK) cascade by membrane-type-1 matrix
metalloproteinase (MT1-MMP). FEBS Lett 507, 231–
236.
31 Langlois S, Gingras D & Be
´
liveau R (2004) Membrane
type 1-matrix metalloproteinase (MT1-MMP) cooper-
ates with sphingosine 1-phosphate to induce endothe-
lial cell migration and morphogenic differentiation.
Blood 103, 3020–3028.
32 Lehti K, Valtanen H, Wickstro
¨
m S, Lohi J &
Keski-Oja J (2000) Regulation of membrane-type-1
matrix metalloproteinase activity by its cytoplasmic
domain. J Biol Chem 275, 15006–15013.
33 Sakamoto T & Seiki M (2009). Cytoplasmic tail of MT1-
MMP regulates macrophage motility independently
from its protease activity. Genes Cells 14, 617–626.
34 Li XY, Ota I, Yana I, Sabeh F & Weiss SJ (2008)
Molecular dissection of the structural machinery
underlying the tissue-invasive activity of membrane
type-1 matrix metalloproteinase. Mol Biol Cell 19,
3221–3233.
35 Hotary K, Allen E, Punturieri A, Yana I & Weiss SJ
(2000) Regulation of cell invasion and morphogenesis
in a three-dimensional type I collagen matrix by
membrane-type matrix metalloproteinases 1, 2, and 3.

J Cell Biol 149, 1309–1323.
G. Murphy and H. Nagase MMPs in the pericellular environment
FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS 11
36 Hotary K, Li XY, Allen E, Stevens SL & Weiss SJ
(2006) A cancer cell metalloprotease triad regulates the
basement membrane transmigration program. Genes
Dev 20, 2673–2686.
37 Gao G, Plaas A, Thompson VP, Jin S, Zuo F &
Sandy JD (2004) ADAMTS4 (aggrecanase-1) activa-
tion on the cell surface involves C-terminal cleavage
by glycosylphosphatidyl inositol-anchored membrane
type 4-matrix metalloproteinase and binding of the
activated proteinase to chondroitin sulfate and hepa-
ran sulfate on syndecan-1. J Biol Chem 279, 10042–
10051.
38 Pilcher BK, Dumin JA, Sudbeck BD, Krane SM,
Welgus HG & Parks WC (1997) The activity of
collagenase-1 is required for keratinocyte migration on
a type I collagen matrix. J Cell Biol 137, 1445–1457.
39 Dumin JA, Dickeson SK, Stricker TP, Bhattacharyya-
Pakrasi M, Roby JD, Santoro SA & Parks WC (2001)
Pro-collagenase-1 (matrix metalloproteinase-1) binds
the a
2
b
1
integrin upon release from keratinocytes
migrating on type I collagen. J Biol Chem 276, 29368–
29374.
40 Ellerbroek SM, Wu YI, Overall CM & Stack MS

(2001) Functional interplay between type I collagen
and cell surface matrix metalloproteinase activity.
J Biol Chem 276, 24833–24842.
41 Wolf K, Mazo I, Leung H, Engelke K, von Andrian
UH, Deryugina EI, Strongin AY, Bro
¨
cker EB & Friedl
P (2003) Compensation mechanism in tumor cell
migration: mesenchymal-amoeboid transition after
blocking of pericellular proteolysis. J Cell Biol 160,
267–277.
42 Ga
´
lvez BG, Matı
´
as-Roma
´
nS,Ya
´
n
˜
ez-Mo
´
M, Sa
´
nchez-
Madrid F & Arroyo AG (2002) ECM regulates MT1-
MMP localization with beta1 or alphavbeta3 integrins
at distinct cell compartments modulating its internali-
zation and activity on human endothelial cells. J Cell

Biol 159, 509–521.
43 Brooks PC, Stro
¨
mblad S, Sanders LC, Von
Schalscha TL, Aimes RT, Stetler-Stevenson WG,
Quigley JP & Cheresh DA (1996) Localization of
matrix metalloproteinase MMP-2 to the surface of
invasive cells by interaction with integrin avb3. Cell
85, 683–693.
44 Brooks PC, Silletti S, Von Schalscha TL,
Friedlander M & Cheresh DA (1998) Disruption of
angiogenesis by PEX, a noncatalytic metalloproteinase
fragment with integrin binding activity. Cell 92, 391–400.
45 Petitclerc E, Stro
¨
mblad S, Von Schalscha TL,
Mitjans F, Piulats J, Montgomery AM, Cheresh DA
& Brooks PC (1999) Integrin alpha(v)beta3 promotes
M21 melanoma growth in human skin by regulating
tumor cell survival. Cancer Res 59, 2724–2730.
46 Deryugina EI, Ratnikov BI, Postnova TI, Rozanov DV
& Strongin AY (2002) Processing of integrin alpha(v)
subunit by membrane type 1 matrix metalloproteinase
stimulates migration of breast carcinoma cells on
vitronectin and enhances tyrosine phosphorylation of
focal adhesion kinase. J Biol Chem 277, 9749–9756.
47 Ga
´
lvez BG, Matı
´

as-Roma
´
n S, Albar JP, Sa
´
nchez-
Madrid F & Arroyo AG (2001) Membrane type
1-matrix metalloproteinase is activated during migration
of human endothelial cells and modulates endothelial
motility and matrix remodeling. J Biol Chem 276,
37491–37500.
48 Iida J, Wilhelmson KL, Ng J, Lee P, Morrison C,
Tam E, Overall CM & McCarthy JB (2007) Cell
surface chondroitin sulfate glycosaminoglycan in
melanoma: role in the activation of pro-MMP-2
(pro-gelatinase A). Biochem J 403, 553–563.
49 Nisato RE, Hosseini G, Sirrenberg C, Butler GS,
Crabbe T, Docherty AJ, Wiesner M, Murphy G,
Overall CM, Goodman SL et al. (2005) Dissecting the
role of matrix metalloproteinases (MMP) and integrin
alpha(v)beta3 in angiogenesis in vitro: absence of
hemopexin C domain bioactivity, but membrane-Type
1-MMP and alpha(v)beta3 are critical. Cancer Res 65,
9377–9387.
50 Mu D, Cambier S, Fjellbirkeland L, Baron JL,
Munger JS, Kawakatsu H, Sheppard D, Broaddus VC
& Nishimura SL (2002) The integrin alpha(v)beta8
mediates epithelial homeostasis through MT1-MMP-
dependent activation of TGF-beta1. J Cell Biol 157,
493–507.
51 Shiomi T, Inoki I, Kataoka F, Ohtsuka T,

Hashimoto G, Nemori R & Okada Y (2005)
Pericellular activation of proMMP-7 (promatrilysin-1)
through interaction with CD151. Lab Invest 85,
1489–1506.
52 Yan
˜
ez-Mo
´
M, Barreiro O, Gonzalo P, Batista A,
Megı
´
as D, Genı
´
s L, Sachs N, Sala-Valde
´
s M, Alonso
MA, Montoya MC et al. (2008) MT1-MMP
collagenolytic activity is regulated through association
with tetraspanin CD151 in primary endothelial cells.
Blood 112, 3217–3226.
53 Takino T, Miyamori H, Kawaguchi N, Uekita T,
Seiki M & Sato H (2003) Tetraspanin CD63 promotes
targeting and lysosomal proteolysis of membrane-type
1 matrix metalloproteinase. Biochem Biophys Res
Commun 304, 160–166.
54 Bourguignon LY, Gunja-Smith Z, Iida N, Zhu HB,
Young LJ, Muller WJ & Cardiff RD (1998) CD44v
3,8-10
is involved in cytoskeleton-mediated tumor cell
migration and matrix metalloproteinase (MMP-9)

association in metastatic breast cancer cells. J Cell
Physiol 176, 206–215.
55 Yu Q & Stamenkovic I (1999) Localization of matrix
metalloproteinase 9 to the cell surface provides a
mechanism for CD44-mediated tumor invasion. Genes
Dev 13, 35–48.
MMPs in the pericellular environment G. Murphy and H. Nagase
12 FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS
56 Lambert E, Bridoux L, Devy J, Dasse
´
E, Sowa ML,
Duca L, Hornebeck W, Martiny L & Petitfre
`
re-
Charpentier E (2009) TIMP-1 binding to proMMP-
9 ⁄ CD44 complex localized at the cell surface promotes
erythroid cell survival. Int J Biochem Cell Biol 41,
1102–1115.
57 Redondo-Mun
˜
oz J, Ugarte-Berzal E, Garcı
´
a-Marco
JA, del Cerro MH, Van den Steen PE, Opdenakker G,
Terol MJ & Garcı
´
a-Pardo A (2008) Alpha4beta1
integrin and 190-kDa CD44v constitute a cell surface
docking complex for gelatinase B ⁄ MMP-9 in chronic
leukemic but not in normal B cells. Blood 112, 169–

178.
58 Redondo-Mun
˜
oz J, Ugarte-Berzal E, Terol MJ, Van
den Steen PE, Herna
´
ndez del Cerro M, Roderfeld M,
Roeb E, Opdenakker G, Garcı
´
a-Marco JA & Garcı
´
a-
Pardo A (2010) Matrix metalloproteinase-9 promotes
chronic lymphocytic leukemia b cell survival through
its hemopexin domain. Cancer Cell 17, 160–172.
59 Kajita M, Itoh Y, Chiba T, Mori H, Okada A, Kinoh
H & Seiki M (2001) Membrane-type 1 matrix metallo-
proteinase cleaves CD44 and promotes cell migration.
J Cell Biol 153, 893–904.
60 Marrero-Diaz R, Bravo-Cordero JJ, Megı
´
as D, Garcı
´
a
MA, Bartolome
´
RA, Teixido J & Montoya MC (2009)
Polarized MT1-MMP-CD44 interaction and CD44
cleavage during cell retraction reveal an essential role
for MT1-MMP in CD44-mediated invasion. Cell Motil

Cytoskeleton 66, 48–61.
61 Yu WH & Woessner JF Jr (2000) Heparan Sulfate
Proteoglycans as Extracellular Docking Molecules for
Matrilysin (Matrix Metalloproteinase 7). J Biol Chem
275, 4183–4191.
62 Yu WH, Woessner JF Jr, McNeish JD & Stamenkovic
I (2002) CD44 anchors the assembly of matrily-
sin ⁄ MMP-7 with heparin-binding epidermal growth
factor precursor and ErbB4 and regulates female
reproductive organ remodeling. Genes Dev 16, 307–
323.
63 Berton A, Selvais C, Lemoine P, Henriet P, Courtoy
PJ, Marbaix E & Emonard H (2007) Binding of matri-
lysin-1 to human epithelial cells promotes its activity.
Cell Mol Life Sci 64, 610–620.
64 Koyama Y, Naruo H, Yoshitomi Y, Munesue S,
Kiyono S, Kusano Y, Hashimoto K, Yokoi T,
Nakanishi H, Shimizu S et al. (2008) Matrix metallo-
proteinase-9 associated with heparan sulfate chains of
GPI-anchored cell surface proteoglycans mediates
motility of murine colon adenocarcinoma cells.
J Biochem 143, 581–592.
65 Herz J & Strickland DK (2001) LRP: a multifunction-
al scavenger and signaling receptor. J Clin Invest 108,
779–784.
66 Yang Z, Strickland DK & Bornstein P (2001) Extracel-
lular Matrix Metalloproteinase 2 levels are regulated
by the Low DensityLlipoprotein-related scavenger
receptor an thrombospondin 2. J Biol Chem 276,
8403–8408.

67 Emonard H, Belton G, Troeberg L, Berton A, Robinet
A, Henriet P, Marbaix E, Kirkegaard K, Patthy L,
Eeckhout Y et al. (2004) Low density lipoprotein
receptor-related protein mediates endocytic clearance
of pro-MMP-2.TIMP-2 complex through a thrombo-
spondin-independent mechanism. J Biol Chem 279,
54944–54951.
68 Hahn-Dantona E, Ruiz JF, Bornstein P & Strickland
DK (2001) The Low Density Lipoprotein Receptor-
related Protein Modulates Levels of Matrix Metallo-
proteinase 9 (MMP-9) by Mediating Its Cellular
Catabolism. J Biol Chem 276, 15498–15503.
69 Barmina OY, Walling HW, Fiacco GJ, Freije JM,
Lo
´
pez-Otı
´
n C, Jeffrey JJ & Partridge NC (1999)
Collagenase-3 binds to a specific receptor and requires
the low density lipoprotein receptor-related protein for
internalization. J Biol Chem 274, 30087–30093.
70 Troeberg L, Fushimi K, Khokha R, Emonard H,
Ghosh P & Nagase H (2008) Calcium pentosan poly-
sulfate is a multifaceted exosite inhibitor of aggrecan-
ases. FASEB J 22 , 3515–3524.
71 Van den Steen PE, Van Aelst I, Hvidberg V, Piccard
H, Fiten P, Jacobsen C, Moestrup SK, Fry S, Royle
L, Wormald MR et al. (2006) The hemopexin and
O-glycosylated domains tune gelatinase B ⁄ MMP-9
bioavailability via inhibition and binding to cargo

receptors. J Biol Chem 281, 18626–18637.
72 Piccard H, Van den Steen PE & Opdenakker G (2007)
Hemopexin domains as multifunctional liganding
modules in matrix metalloproteinases and other
proteins. J Leukoc Biol 81, 870–892.
73 Monferran S, Paupert J, Dauvillier S, Salles B &
Muller C (2004) The membrane form of the DNA
repair protein Ku interacts at the cell surface with
metalloproteinase 9. Embo J 23, 3758–3768.
74 Biswas C, Zhang Y, DeCastro R, Guo H,
Nakamura T, Kataoka H & Nabeshima K (1995) The
human tumor cell-derived collagenase stimulatory
factor (renamed EMMPRIN) is a member of the
immunoglobulin superfamily. Cancer Res 55, 434–439.
75 Nabeshima K, Iwasaki H, Koga K, Hojo H, Suzumiya
J & Kikuchi M (2006) Emmprin (basigin ⁄ CD147):
matrix metalloproteinase modulator and multifunc-
tional cell recognition molecule that plays a critical
role in cancer progression. Pathol Int 56, 359–367.
76 Guo HM, Li RS, Zucker S & Toole BP (2000) EMM-
PRIN (CD147), an inducer of matrix metalloprotein-
ase synthesis, also binds interstitial collagenase to the
tumor cell surface. Cancer Res 60, 888–891.
77 Noguchi Y, Sato T, Hirata M, Hara T, Ohama K &
Ito A (2003) Identification and characterization of
extracellular matrix metalloproteinase inducer in
G. Murphy and H. Nagase MMPs in the pericellular environment
FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS 13
human endometrium during the menstrual cycle in vivo
and in vitro. J Clin Endocrinol Metab 88, 6063–6072.

78 Isacke CM, van der GP, Hunter T & Trowbridge IS
(1990) p180, a novel recycling transmembrane glyco-
protein with restricted cell type expression. Mol Cell
Biol 10, 2606–2618.
79 Wu K, Yuan J & Lasky LA (1996) Characterization
of a novel member of the macrophage mannose recep-
tor type C lectin family. J Biol Chem 271, 21323–
21330.
80 Behrendt N, Jensen ON, Engelholm LH, Mortz E,
Mann M & Dano K (2000) A urokinase receptor-asso-
ciated protein with specific collagen binding properties.
J Biol Chem 275, 1993–2002.
81 Leitinger B & Hohenester E (2007) Mammalian colla-
gen receptors. Matrix Biol 26, 146–155.
82 Lee H, Overall CM, McCulloch CA & Sodek J (2006)
A critical role for the membrane-type 1 matrix metal-
loproteinase in collagen phagocytosis. Mol Biol Cell
17, 4812–4826.
83 Messaritou G, East L, Roghi C, Isacke CM &
Yarwood H (2009) Membrane type-1 matrix metallo-
proteinase activity is regulated by the endocytic
collagen receptor Endo180. J Cell Sci 122, 4042–4048.
84 Yamamoto K, Higashi S, Kioi M, Tsunezumi J,
Honke K & Miyazaki K (2006) Binding of active
matrilysin to cell surface cholesterol sulfate is essential
for its membrane-associated proteolytic action and
induction of homotypic cell adhesion. J Biol Chem
281, 9170–9180.
85 Higashi S, Oeda M, Yamamoto K & Miyazaki K
(2008) Identification of amino acid residues of matrix

metalloproteinase-7 essential for binding to cholesterol
sulfate. J Biol Chem 283, 35735–35744.
86 Xu X, Chen Z, Wang Y, Yamada Y & Steffensen B
(2005) Functional basis for the overlap in ligand inter-
actions and substrate specificities of matrix metallopro-
teinases-9 and -2. Biochem J 392, 127–134.
87 Freise C, Erben U, Muche M, Farndale R,
Zeitz M, Somasundaram R & Ruehl M (2009) The
alpha 2 chain of collagen type VI sequesters latent
proforms of matrix-metalloproteinases and modulates
their activation and activity. Matrix Biol 28, 480–
489.
88 Olson MW, Toth M, Gervasi DC, Sado Y,
Ninomiya Y & Fridman R (1998) High affinity binding
of latent matrix metalloproteinase-9 to the a2(IV) chain
of collagen IV. J Biol Chem 273, 10672–10681.
89 Owen CA, Hu Z, Barrick B & Shapiro SD (2003)
Inducible expression of tissue inhibitor of metallopro-
teinases-resistant matrix metalloproteinase-9 on the cell
surface of neutrophils. Am J Respir Cell Mol Biol 29,
283–294.
90 Bannikov GA, Karelina TV, Collier IE, Marmer BL &
Goldberg GI (2002) Substrate binding of gelatinase B
induces its enzymatic activity in the presence of intact
propeptide. J Biol Chem 277, 16022–16027.
91 Jain A, Fisher LW & Fedarko NS (2008) Bone sialo-
protein binding to matrix metalloproteinase-2 alters
enzyme inhibition kinetics. Biochemistry 47, 5986–
5995.
92 Zhang L, Yang M, Yang D, Cavey G, Davidson P &

Gibson G (2010) Molecular interactions of MMP-13
C-terminal domain with chondrocyte proteins. Connect
Tissue Res 51, 230–239.
93 Orgel JP, Irving TC, Miller A & Wess TJ (2006)
Microfibrillar structure of type I collagen in situ. Proc
Natl Acad Sci USA 103, 9001–9005.
94 Perumal S, Antipova O & Orgel JP (2008) Collagen
fibril architecture, domain organization, and triple-heli-
cal conformation govern its proteolysis. Proc Natl
Acad Sci USA
105, 2824–2829.
95 Saffarian S, Collier IE, Marmer BL, Elson EL &
Goldberg G (2004) Interstitial collagenase is a
Brownian ratchet driven by proteolysis of collagen.
Science 306, 108–111.
96 Sweeney SM, Orgel JP, Fertala A, McAuliffe JD,
Turner KR, Di Lullo GA, Chen S, Antipova O,
Perumal S, Ala-Kokko L et al. (2008) Candidate cell
and matrix interaction domains on the collagen fibril,
the predominant protein of vertebrates. J Biol Chem
283, 21187–21197.
97 Gross J & Lapiere CM (1962) Collagenolytic activity
in amphibian tissues: a tissue culture assay. Proc Natl
Acad Sci USA 48, 1014–1022.
98 Mauch S, Kolb C, Kolb B, Sadowski T & Sedlacek R
(2002) Matrix metalloproteinase-19 is expressed in
myeloid cells in an adhesion-dependent manner and
associates with the cell surface. J Immunol 168, 1244–
1251.
99 Overall CM & Blobel CP (2007) In search of partners:

linking extracellular proteases to substrates. Nat Rev
Mol Cell Biol 8, 245–257.
100 Tomari T, Koshikawa N, Uematsu T, Shinkawa T,
Hoshino D, Egawa N, Isobe T & Seiki M (2009) High
throughput analysis of proteins associating with a
proinvasive MT1-MMP in human malignant
melanoma A375 cells. Cancer Sci 100, 1284–1290.
101 Stricker TP, Dumin JA, Dickeson SK, Chung L,
Nagase H, Parks WC & Santoro SA (2001) Structural
analysis of the alpha 2 Integrin I domain ⁄ procollagen-
ase-1 (matrix metalloproteinase-1) interaction. J Biol
Chem 276, 29375–29381.
102 Patterson ML, Atkinson SJ, Kna
¨
uper V & Murphy G
(2001) Specific collagenolysis by gelatinase A, MMP-2,
is determined by the hemopexin domain and not the
fibronectin-like domain. FEBS Lett 503, 158–162.
103 Stefanidakis M, Bjorklund M, Ihanus E,
Gahmberg CG & Koivunen E (2003) Identification of
a negatively charged peptide motif within the catalytic
MMPs in the pericellular environment G. Murphy and H. Nagase
14 FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS
domain of progelatinases that mediates binding to
leukocyte beta 2 integrins. J Biol Chem 278, 34674–
34684.
104 Allan JA, Hembry RM, Angal S, Reynolds JJ &
Murphy G (1991) Binding of latent and high M
r
active

forms of stromelysin to collagen is mediated by the
C-terminal domain. J Cell Sci 99, 789–795.
105 Ra HJ, Harju-Baker S, Zhang F, Linhardt RJ,
Wilson CL & Parks WC (2009) Control of promatrily-
sin (MMP7) activation and substrate-specific activity
by sulfated glycosaminoglycans. J Biol Chem 284 ,
27924–27932.
106 Owen CA, Hu Z, Lo
´
pez-Otı
´
n C & Shapiro SD (2004)
Membrane-bound matrix metalloproteinase-8 on
activated polymorphonuclear cells is a potent, tissue
inhibitor of metalloproteinase-resistant collagenase and
serpinase. J Immunol 172, 7791–7803.
107 Bjorklund M, Heikkila P & Koivunen E (2004) Peptide
inhibition of catalytic and noncatalytic activities of
matrix metalloproteinase-9 blocks tumor cell migration
and invasion. J Biol Chem 279, 29589–29597.
108 Suenaga N, Mori H, Itoh Y & Seiki M (2005) CD44
binding through the hemopexin-like domain is critical
for its shedding by membrane-type 1 matrix metallo-
proteinase. Oncogene 24, 859–868.
109 Iida J, Pei D, Kang T, Simpson MA, Herlyn M,
Furcht LT & McCarthy JB (2001) Melanoma
chondroitin sulfate proteoglycan regulates matrix me-
talloproteinase-dependent human melanoma invasion
into type I collagen. J Biol Chem 276, 18786–18794.
G. Murphy and H. Nagase MMPs in the pericellular environment

FEBS Journal 278 (2011) 2–15 ª 2010 The Authors Journal compilation ª 2010 FEBS 15

×