You are on page 1of 26

Periodontology 2000, Vol.

24, 2000, 127–152 Copyright C Munksgaard 2000


Printed in Denmark ¡ All rights reserved
PERIODONTOLOGY 2000
ISSN 0906-6713

Cell biology of
gingival wound healing
L ARI H ÄKKINEN, V ELI- J UKKA U ITTO & H ANNU L ARJAVA

An overview extracellular molecules and their expression using in


situ hybridization. In addition, transgenic animals
Wound healing is a critical process for the organism. provide information how wound healing is altered in
The attempt of this chapter is to critically summarize the absence or following overexpression of a known
the recent advancements in the wound-healing pro- molecule (153). Many of these studies have provided
cess. Because of the excessive amount of infor- new critical information but others have resulted in
mation (search with the key words wound healing confusing results, such as showing no alterations
produced 2052 citations within the last 12 months), even though a molecule has been thought to be criti-
we will focus on two major events in wound healing, cally important for wound healing. It is believed that
namely re-epithelialization and granulation tissue in these cases there are other related proteins that
formation, leaving out many other key events such can provide similar functions and compensate for
as clot formation and angiogenesis. At the end, we the absence of the studied molecule. Wound healing
will also discuss the special features of wound heal- has become well protected during evolution because
ing in oral cavity. It is commonly stated that oral of its critical importance. Numerous molecules ap-
wounds heal better than other wounds such as der- pear to overlap each other’s functions. During
mal wounds. We will explore evidence supporting wound healing, several molecules that are usually
this assumption. present only during embryonal development are
Most of the data presented in this chapter refer to found in the granulation tissue. Epithelial cells start
studies with gingival and dermal full-thickness to express extracellular matrix receptors that are not
wounds and no special attempts have been made normally present in the resting epithelium. Fibro-
to focus on the healing in tooth-gingiva interphase blastic cells with a special phenotype are found in
because only a few studies are available to report the healing granulation tissue. Where they come
special molecular events in humans. In addition, from is still largery unknown but possible sources
several previous articles have addressed periodontal will be discussed in this chapter. Expression of vari-
wound healing and regeneration in great detail (114, ous proteins by the resident cells is also influenced
276). It is reasonable to assume, however, that the by the new environment. For example, in fibroblasts
basic cell biological events of wound healing will fol- the expression of hundreds of genes is altered by the
low the same principles at the tooth-gingiva inter- exposure to serum alone. Proteolytic enzymes re-
phase, at least at the supracrestal locations. We will lease growth factors from the wounded basement
also try to make several back-and-forth citations be- membrane and connective tissue matrix. Matrix
tween in vivo and in vitro studies. This is necessary degradation produces biologically active peptides
because functional studies have been difficult to from the matrix proteins that could have specific
perform with animal models of wound healing in a functions in tissue repair. Clarification of the com-
manner that would allow meaningful conclusions to plex interplay between new matrix, growth factors,
be drawn. In vitro studies with cultured keratino- matrix degradation products and cells during wound
cytes and fibroblasts are still the only way to explore healing will be a challenging task. This review fo-
functions of various molecules. Recent molecular bi- cuses on re-epithelialization and granulation tissue
ology techniques have made it possible to eliminate formation with the special emphasis on the inte-
the expression of molecules of interest and investi- grins, since cell adhesion serves as a foundation for
gate how the function is changed consequently. In cell migration, matrix turnover and differentiation
vivo studies have dealt with immunolocalization of during wound healing.

127
Häkkinen et al.

Integrins being used for designing new molecules that have


high affinity to the binding site but are irrelevant to
Integrins play key roles in re-epithelialization and the original peptide sequence. Commercial products
granulation tissue formation during wound healing are already available for cardiovascular applications
through their function in cell adhesion and sig- (146). Several other compounds are in clinical trials
naling. A brief overview is, therefore, presented de- and many other similar products will follow and may
scribing the structure and function of integrins that be used for periodontal applications in the future.
we hope will help the reader to better understand The number of proteins known to associate with
how they participate in the regulation of wound integrins is rapidly increasing (96). Integrin associ-
healing. Integrins are cell surface–associated dimeric ated protein, transmembrane-4 superfamily, growth
glycoproteins that function as cell-to-extracellular factor receptors and urokinase-type plasminogen ac-
matrix adhesion receptors (4, 16, 160). Through tivator receptor appear to have a regulatory function
binding to the extracellular matrix proteins integrins on integrins. Growth factor receptors accumulate in
mediate information transfer from the extracellular the same structures as integrins and regulate inte-
matrix to the cell interior leading to alterations in grin functions (Fig. 1) (161). In the context of wound
cell functions and ultimately in cell behaviour (100, healing, the synergy between integrin and growth
160). Integrins are known to play an important role factor receptors is probably a key process in the
in regulating a wide range of cell functions during regulation of cell proliferation (100). Integrins avb1
growth, development, differentiation, and immune and avb6 can also bind growth factors such as trans-
response (4). Integrins are composed of a single a forming growth factor b1 through its latency-acti-
and a single b subunit that are non-covalently linked vated peptide that contains an Arg-Gly-Asp (RGD)
to each other. At least 17 different a and 8 b subunits peptide sequence (166, 167). It has recently been
are currently known. These subunits can variously shown that epithelial cells can bind and also activate
combine to form more than 23 different cell surface transforming growth factor b1 through avb6 integrin
receptors that have distinct ligand-binding speci- (Fig. 1); this mechanism may play an important role
ficities. Both a and b subunits are transmembrane in the connective tissue bridge formation under-
glycoproteins that cooperate in integrin binding to neath the epithelium that has covered the wound. It
ligands. Ligand binding causes clustering of the re- is evident that the epithelium has a much more ac-
ceptors, which leads to cytoskeletal organization and tive role in the regulation of connective tissue forma-
signaling (Fig. 1) (160). Integrin-binding sites are tar- tion than previously assumed.
gets for therapeutic applications aiming at blocking
integrin function. Domain crystal structures are
Epithelial wound healing
Re-epithelialization

Wound healing is a complex phenomenon that in-


volves series of controlled events including the for-
mation of a provisional extracellular matrix that is
mainly composed of fibrin, fibronectin and vitronec-
tin and the migration of epithelial cells from the edg-
es of the wound (33, 108). After the epithelium has
been disrupted by tissue injury, re-epithelialization
must occur as rapidly as possible in order to re-es-
tablish tissue integrity. Keratinocytes start moving
into the defect about 24 hours after the injury (277).
Epithelial cells from residual epithelial structures
Fig. 1. Integrins and cell signalling. Integrins mediate cell dissolve their hemidesmosomal connections and de-
adhesion to extracellular matrix proteins (ECM) leading tach from basement membrane and move quickly
to organization of cytoskeleton and signaling. Integrins across the wound defect. Later, as the re-epithelializ-
also collaborate with growth factors in regulation of cell ation proceeds, the proliferation of keratinocytes
growth. Some integrins can also directly activate growth
factors such as transforming growth factor b1 (TGFb1).
that feed the advancing epithelial edge becomes
Proteolytic enzymes are also found in cell adhesion sites more important. It is still somewhat unclear which
allowing cells to detach and subsequently migrate. cells in the epithelium first move into the wound.

128
Cell biology of gingival wound healing

There is some evidence that the suprabasal keratino- also be necessary. Migrating keratinocytes express
cytes are the first migratory cells sliding over the matrix metalloprotease-9 (type IV collagenase), ma-
basal keratinocytes. Epithelial cell migration on the trix metalloprotease-1 (interstitial collagenase) and
exposed connective tissue matrix underneath the matrix metalloprotease-10 (stromelysin), which all
fibrin-fibronectin clot has been commonly de- may be required if the cells encounter the exposed
scribed. In small gingival wounds (Fig. 2), however, matrix (207, 210, 266). Blocking matrix metalloprote-
the keratinocytes cut their way directly through the ase activity prevents keratinocyte migration into the
clot and may not interact with the connective tissue wounds in cell culture (147). This proteolytic modu-
matrix at all (132). Migrating keratinocytes are highly lation of the matrix underneath migrating cells must
phagocytic, allowing them to penetrate through be well controlled, since overexpression of matrix
tissue debris or the clot (277). Degradation of the fi- metalloproteases is a common finding in nonhealing
brin clot appears to be critical for wound healing, chronic wounds (187). In chronic inflammation, ma-
since wounds in animals that lack the plasminogen trix metalloproteases such as matrix metalloprotea-
gene do not re-epithelialize (199). It seems likely that se-9 produced by keratinocytes may bind to the cell
integrins play a role in the fibrin clot removal. Mi- surface and into the extracellular matrix resulting in
grating cells must be able to focalize the proteolysis a delayed clearance (147). Thus, regulation of matrix
into the leading edge of epithelium (227). This could metalloproteases and their inhibitors must be well
be done by activation of proteolytic enzymes at spe- balanced for normal wound healing (266).
cific sites at the cell membrane. It has been found During wound healing, keratinocytes function to
that urokinase type plasminogen activator receptor rapidly cover the exposed connective tissue (236,
is able to associate with integrins (283). This is an 277). This process depends upon a variety of interac-
example how a cell is able to focalize fibrinolysis by tions between cells and the extracellular matrix. As
plasmin and promote subsequent migration by inte- the basal keratinocytes at the wound margin are ex-
grins. Modulation of other matrix componets may posed to the new provisional matrix, the phenotype

Fig. 2. Stages of healing of full thickness gingival wounds (about 3 mm deep and 2 mm wide). FC: fibrin clot; GT:
granulation tissue.

129
Häkkinen et al.

of the cells is changed from stationary to migratory Laminin-5 can be proteolytically modified (see be-
(176). In this process, keratinocytes detach them- low) to produce different molecules that function
selves from the basement membrane, migrate lat- either as a nucleator of the hemidesmosomes (stop-
erally into the wound bed and finally regenerate the ping signal) or as a promoter of migration. Laminin-
basement membrane (124). In mucosa and in skin, 5 seems to be the nucleator of a6b4 integrin and
the migration seems to involve similar patterns, al- therefore the nucleator of basement membrane or-
though the source of migrating epithelial cells is dif- ganization (110). Migrating keratinocytes produce
ferent. In the mucosa, the basal layers of the wound other extracellular matrix molecules that they could
edge epithelium serves as a major source of the kera- use to support their own migration. Fibronectin-EII-
tinocytes migrating into the wound area. In the skin, IA but not EIIIB (see below) appears to be present
however, epithelial cells arise not only from the underneath the migrating keratinocytes in vivo (Fig.
wound edge but also from hair follicles and sweat 3) (Häkkinen et al., unpublished). In addition, kera-
glands. During wound healing, most of the compo- tinocytes in culture deposit fibronectin-EIIIA while
nents of the basement membrane zone such as type they migrate on substrates coated with serum
IV and VII collagens, laminin-1 and heparan sulfate fibronectin (Koivisto et al., unpublished). Tenascin-
proteoglycan are missing underneath migrating C large and small variants are also found underneath
keratinocytes (132, 178). Laminin-5, however, ap- the migrating keratinocytes. As we will discuss else-
pears to be always deposited against the wound bed where in this chapter, tenascin-C may function as a
matrix by keratinocytes during migration (Fig. 3) modulator of cell adhesion to other matrix compo-
(132). The role of laminin-5 appears to be critical nents such as fibronectin. It appears obvious, there-
even for healing of the blister wounds where lamina fore, that keratinocytes themselves are capable of
densa remains intact (110). It is also the first base- making extracellular matrix that they can use to sup-
ment membrane component found against the con- port or modulate their own migration if the pro-
nective tissue in experimental cyst formation that re- visional matrix is not permissive to migration. One
sembles wound healing (110). Keratinocytes show could question whether keratinocytes in healing
poor migration in chronic aphthous ulcers and also wounds migrate on the provisional matrix at all.
fail to deposit a proper laminin-5 rich matrix (196). Some of the migration could be intraepithelial, that

Fig. 3. Keratinocyte integrins and extracellular matrix pro- (LM5: laminin-5; TN: tenascin-C; FN ED-A: fibronectin
teins during re-epithelialization. A. Histological represen- isoform EIIIA) while they migrate through wound pro-
tation of a 3-day-old wound. B. Schematic view of the visional matrix. FC: fibrin clot; CT: connective tissue; E:
same wound. Keratinocytes express a number of integrin epithelium. πππ refers to the strong immunoreaction at
receptors (a2b1 to a6b4) and extracellular matrix proteins this stage of healing.

130
Cell biology of gingival wound healing

Table 1. Epithelial integrins and their ligands during wound healing


Integrin Ligand Ligand source
a2b1 Collagen Connective tissue
Tenascin (?) Keratinocytes, granulation tissue
a3b1, a6b4 Laminin-5 Keratinocytes
a5b1, avb1 Serum fibronectin Serum
Fibronectin EIII/A Keratinocytes
a9b1 Tenascin-C Keratinocytes, granulation tissue
avb5 Vitronectin Serum, connective tissue
avb6 Fibronectin EIIIA, fibronectin EIIIB Keratinocytes, granulation tissue
Transforming growth factor b1 Keratinocytes, macrophages, connective tissue cells

is, suprabasal keratinocytes sliding on basal cells at nents of the basement membrane zone (236). Sig-
the leading edge. After reaching the provisional ma- nificant portion of the basement membrane compo-
trix, migrating cells could stop moving and become nents are, however, synthesized by the wound
basal stationary keratinocytes. Two models for epi- fibroblasts (65). It is obvious that the cross-talk be-
thelial sheet migration in wounds have been pro- tween the wound keratinocytes and fibroblasts is
posed (236). In the sliding model the cells at the crucial during the reorganization of the basement
margin are active and pull the cells behind them. In membrane zone.
a leap frog model, the migrating cells become non-
motile when they adhere to the provisional wound
matrix. Then the cells behind the leading edge crawl Extracellular matrix interactions of keratinocytes
during re-epithelialization
over the newly attached keratinocytes. The exact
mechanism by which multilayered epithelium mi- As the epithelial cells undergo major environmental
grates remains to be discovered, but it is possible changes during the wound healing, they have to ad-
that all three mechanisms described above could just their cell surface receptors for the new demands.
function depending on the defect and the state of During wound healing keratinocytes have to migrate
the healing. Embryonic re-epithelialization appears on the provisional matrix different from their
to occur through a different mechanism. Epidermal stationary matrix. This requires a change of express-
cells do not crawl by extending lamellopodia into the ion levels and distribution of old integrins and ex-
wound as they do during, for example, gingival pression of some totally new integrins (Table 1, Fig.
wound healing (132). They rather use the actin 3) (131). As discussed throughout this chapter,
filament cables to pull wound edges together (154). fibronectin is a critical early component of the clot
In small gingival wounds (2–3 mm distance be- and the forming granulation tissue. Initially, the
tween the wound edges), the formation of new base- blood clot contains plasma fibronectin, which is
ment membrane starts when the migrating epithelial later replaced by cellular fibronectin produced by
sheets have reached each other and fused (132). The keratinocytes, fibroblasts and macrophages (33).
nucleation of the basement membrane appears to Fibronectin can vary structurally in a tissue specific
occur at multiple sites at the same time. It has been manner by alternative splicing of three regions,
reported that in epidermal wounds the deposition of namely EIIIA, EIIIB and V(IIICS) (284). Expression of
basement membrane starts from the wound margin the alternatively spliced isoforms of cellular
in a zipper-like fashion (35). Whether there is a real fibronectin containing either the EIIIA or EIIIB mod-
difference in the sequence of basement membrane ules, which are omitted from the plasma fibronectin,
organization between gingival and epidermal heal- are upregulated specifically during embryonic devel-
ing has not been shown. The reorganization of the opment and upon wounding (Häkkinen et al., un-
basement membrane is complete at 4 weeks at published). As discussed above, EIIIA fibronectin is
which time the localization of all basement mem- expressed by migrating keratinocytes in wounds
brane components such as type IV and VII collagens, (Häkkinen et al., unpublished). The EIIIB fibronectin
laminin-1 and heparan sulfate proteoglycan appear that is expressed in embryonal tissues but not nor-
normal (Häkkinen et al., unpublished). Keratinocytes mally in adult connective tissue is strongly upregu-
have been shown to synthesize these main compo- lated during granulation tissue formation (Häkkinen

131
Häkkinen et al.

et al., unpublished, see below). Plasma fibronectin the basal keratinocytes, suggesting that a3b1 inte-
and the isoforms containing the EIIIA or EIIIB mod- grin collaborates with a6b4 integrin in laminin-5
ules may have different properties in cell adhesion binding and they may be functionally interchange-
and migration during wound repair. Human wound able in some circumstances. Integrin a6b4 is present
keratinocytes express two major receptors that are in migratory epithelial cells although hemidesmo-
able to bind to fibronectins, namely a5b1 and avb6 somes are absent (132). One possible scenario could
integrins (Table 1) (91, 132). The classical fibronectin be connected to the functions of laminin-5 during
binding receptor is a5b1 integrin that binds to the wound healing. Intact laminin-5 appears to be a mo-
RGD sequence of the fibronectin molecule (1, 229). tility factor for keratinocytes, but when proteo-
Integrin a5b1 is considered to be a specialized recep- lytically processed it starts to function as the nu-
tor for fibronectin and it mediates fibronectin-matrix cleator of hemidesmosomes and therefore promotes
assembly, cell adhesion and migration on fibronec- the formation of basement membrane structures
tin (1). Another fibronectin RGD site binding recep- (78). This is a good example demonstrating how
tor in epithelial cells is the avb6 integrin (17). Inte- complex the wound-healing process is, since simple
grin avb6 is expressed exclusively in epithelial cells, proteolytic cleavage can totally reverse the function
and it functions as a receptor for the extracellular of a protein important for the cell migration process.
matrix proteins fibronectin (17, 270) and tenascin Expression of b1 integrins is strongly upregulated
(191). Neither of the main fibronectin receptors, in keratinocytes during wound healing (20, 73, 97,
a5b1 or avb6 integrin, appears to be present in kera- 108, 132). Both a2b1 and a3b1 integrins seem to be
tinocytes residing in the resting epithelium, but the equally expressed. Integrin a3b1 is known to be able
expression of both of these integrins can be induced to bind both fibronectin and laminin-5, both of
by wounding or placing epithelial cells in cell culture which are present in the provisional matrix (19, 132)
(Fig. 3). These two fibronectin receptors appear in although its role as a fibronection receptor in kera-
the wound keratinocytes at different times, a5b1 tinocytes is doubtful. Integrin a2b1 is known to be
during early migration and avb6 during reorganiza- able to bind many types of collagens (Table 1), such
tion of the basement membrane zone suggesting as type I, III, V and VI that serve as a substrate for
that their function may be different (91, Häkkinen migrating keratinocytes. Direct interactions of kera-
et al., unpublished). As mentioned earlier, epithelial tinocytes with type IV collagen are rare but may oc-
integrin avb6 may play a central role in the reorgan- cur, for example, during healing of blister wounds,
ization of the basement membrane zone (Fig. 4) as it in which case keratinocytes migrate on the compo-
is expressed by wound keratinocytes after epithelial nents of lamina densa. Keratinocytes are more likely
sheets have joined and because it is able to activate to interact with fibrillar collagens, such as types I,
transforming growth factor b (167). In culture, kera- III and V collagens during healing of full-thickness
tinocytes can switch between avb6 and a5b1 inte- wounds. In this case, interaction of keratinocytes
grins in fibronectin binding if one receptor is un- with collagens is mediated via a2b1 integrin. Inte-
functional (118). grins are also involved in the regulation of extracellu-
Laminin-5 is found in the basement membrane lar matrix degradation. Cells in the tissue adapt to
of skin and other epithelial tissues and serves as a their changing environment and sense alterations in
component of anchoring filaments that span the matrix through integrins on the cell surface. Rec-
through the basement membrane (19, 203). The ognition of an altered matrix by integrins lead
extracellular domain of a6b4 integrin interacts with changes in gene expression. For example, epithelial
laminin-5 (173, 203). This integrin is known to link and connective tissue cells attach to collagen by
the basal keratinocytes to the underlying basement a2b1 integrin which process induces the expression
membrane, and this link mainly relies on the inter- of collagenase (matrix metalloproteinase-1) that can
action with laminin-5 in the anchoring filaments. cleave the exposed collagen matrix and facilitate the
The functional importance of a6b4 integrin is sub- migration of wound keratinocytes (186, 187, 278).
stantiated by experiments in knockout mice that lack Vitronectin is an abundant protein present in
either a6 or b4 integrin and cannot form hemides- blood plasma, extracellular matrices and fibrin clots.
mosomes, which results in blistering of skin. Lamin- Vitronectin is known to be an active cell adhesion
in-5 is also recognized by a3b1 integrin (19). Trans- mediator as well as playing a role in cell migration
genic mice lacking a3 integrin also develop localized and invasion (190). The avb1, avb3, avIIb3 and avb5
blistering of the skin (49). In mice and man, a3b1 integrins are all vitronectin receptors binding to the
integrin is occasionally found at the basal surface of RGD cell attachment sequence of the protein (24).

132
Cell biology of gingival wound healing

Fig. 4. Hypothetical role of keratinocyte avb6 integrin in thelium that cover the granulation tissue (GT) express
the regulation of granulation tissue formation. A. Fibrin avb6 integrin. C. Schematic representation of the putative
staining of a 7-day-old gingival wound. Arrowheads mark role of avb6 integrin in wound healing. TGFb1: trans-
the wound area. B. Immunolocalization of avb6 integrin forming growth factor b1.
in a 7-day-old wound. Basal keratinocytes of fused epi-

Migrating keratinocytes in porcine epidermis have ecules that can replace its function and compensate
been reported to express the avb5 integrin (73). The for its absence. Tenascin-C is found in developing
avb5 integrin is present in cultured keratinocytes brain and in mesenchyme associated with epithelial-
and has been shown to mediate keratinocyte mi- mesenchymal interactions (28, 247). In adult tissues
gration on vitronectin (116). Vitronectin is present in tenascin-C is found in the connective tissue under-
the provisional wound bed matrix and it is, therefore, neath epithelium (138), but its expression is strongly
reasonable to assume that an avb5-vitronectin inter- upregulated during wound healing (31, 144, 275). As
action can exist during wound healing (Table 1) (36). mentioned above, the large and small tenascin-C
Tenascins are a family of large extracellular matrix variants are found under migrating keratinocytes,
proteins whose expression is tightly regulated, mak- suggesting their active role in re-epithelialization.
ing this protein particularly interesting. The express- Whether tenascins modulate keratinocyte adhesion
ion of tenascin is closely associated with morpho- to other matrix components or support directly kera-
genetic events, including embryonic induction and tinocyte adhesion remains to be shown. Several
migration, wound healing and tumorigenesis (28, keratinocyte integrins have been found to bind tena-
122, 275). Tenascin-C knockout mice develop nor- scin-C, namely a2b1, avb6 and a9b1 integrins (14,
mally (208), suggesting that there are other mol- 191, 290, 291). Integrin a9b1 in actively migrating,

133
Häkkinen et al.

and avb6 integrin in nonmigrating wound keratino- HaCaT keratinocytes, transforming growth factor b1
cytes, colocalize in the same area as tenascins (Table and 2 specifically stimulate the expression of avb6
1) (Häkkinen et al., unpublished). These two epi- integrin, and promote both haptotactic and epi-
thelial integrins may, therefore, have a different role thelial sheet migration (118). As we have discussed
in cell signaling caused by tenascin-C variants. Up- earlier, the most important role for avb6 integrin
regulation of a9b1 and tenascin-C is also observed may be the activation of transforming growth factor
during corneal wound healing (239). b1 that then promotes the formation of the connec-
tive tissue bridge formation under the joined wound
epithelium. The first collagen fibrils are, indeed, laid
Transforming growth factor b and down under the epithelium in areas where the ex-
re-epithelialization
pression of avb6 is strong and transforming growth
In wounds, specific regulatory signals are required factor b1 is present in active form. Transforming
for normal repair. Currently it is believed that trans- growth factor b1 may also stimulate the proliferation
forming growth factors-b have a central role in the of wound keratinocytes at the wound margins in-
wound healing process. Transforming growth factors directly by inducing the expression of other polypep-
b are a family of polypeptides that have multiple tide growth factors, such as platelet-derived growth
regulatory actions in cell growth, differentiation, and factor (3, 22).
developmental processes (155, 234, 244). Three In addition to transforming growth factors b,
highly homologous transforming growth factor-b many other growth factors regulate wound healing.
genes have been identified in mammals, represent- This review will not try to list all possible factors in-
ing transforming growth factor b1, transforming volved in re-epithelialization and the readers are ad-
growth factor b2 and transforming growth factor b3 vised to review other sources for the roles of platelet-
polypeptides. Human keratinocytes of intact skin ex- derived growth factor, epidermal growth factor, kera-
press transforming growth factor b3 (216). This tinocyte growth factor, hepatocyte growth factor and
growth factor seems to play an important role in epi- others in wound healing.
dermal maintenance. In animal studies, only small
amounts of transforming growth factor b2 and trans-
forming growth factor b3 messenger RNA have been
Connective tissue repair
found in keratinocytes of intact dermis (215). All
Activation of fibroblasts
transforming growth factor b isoforms are found in
healing wounds of animals (111, 137). However, The resolution of a tissue defect after wounding re-
Schmid et al. (216) did not find any detectable levels quires not only re-epithelialization but also prolifer-
of transforming growth factor b2 messenger RNA in ation and migration of fibroblasts into the wound
human wound keratinocytes. Wound fibroblasts and bed where they participate in the formation of
macrophages are known to express both trans- granulation tissue and synthesize, deposit and reor-
forming growth factor b1 and transforming growth ganize various extracellular matrix molecules.
factor b2 in cells adjacent to the wound (175, 267). Wound repair involves phenotypic change of fibro-
Transforming growth factor b1 is the major isoform blasts from quiescent to proliferating cells, and sub-
in wound keratinocytes, and induction of trans- sequently to migratory, and then to stationary matrix
forming growth factor b1 in migrating keratinocytes producing and contractile cells. In normal connec-
is crucial for the successful re-epithelialization of tive tissue, fibroblasts reside in a quiescent state and
skin wounds. In vitro studies have shown that trans- have a slow proliferation rate and metabolic activity.
forming growth factor b3 inhibits the growth of pri- Upon wounding, fibroblasts become activated and
mary human keratinocytes, while transforming change their gene expression. This is supported by a
growth factor b1 seems to stimulate keratinocyte recent finding with quiescent fibroblasts that show a
motility by switching the cells from the differentiat- rapid change in expression of hundreds of genes
ing to regenerative phenotype (152) and by inducing when stimulated by serum. Interestingly, many of
their production of fibronectin (172) and laminin-5 the serum-activated genes are known to be involved
(110). In cultured human keratinocytes, trans- in the physiology of wound repair, including control
forming growth factor b1 has also been shown to in- of cell cycle and proliferation, coagulation and
crease the levels of messenger RNA for some inte- hemostasis, inflammation, angiogenesis, tissue re-
grins, such as a5, av and b5, that may facilitate mi- modeling, cytoskeletal reorganization and re-epi-
gration of wound keratinocytes (73, 292). In cultured thelialization (107). This indicates that exposure of

134
Cell biology of gingival wound healing

fibroblasts to serum that is present in the blood clot 185, 188, 219, 225) suggesting that signals initiated
in the wound initiates not only a rapid general by wounding may stimulate certain subpopulations
stimulus for cell proliferation but also a more speci- to enter the wound space. On the other hand, there
fic gene expression that controls function of other is also evidence that some of the fibroblasts that
cells involved in inflammation, angiogenesis and re- have migrated into wound actually change pheno-
epithelialization. Therefore, it is likely that fibro- type and differentiate into myofibroblasts (see
blasts play a more important role in the physiology below).
of wound repair than has been previously realized Progenitor fibroblast populations have been iden-
(107). tified in wounds for decades using autoradiographic
In addition to serum stimulation, cell adhesion to labeling techniques. Based on these studies, the
different matrix molecules can also alter gene ex- main source of wound fibroblasts seems to be the
pression and protein synthesis (4, 120, 121, 143). Cell surrounding connective tissue. Typically, 2 or 3 days
adhesion also modulates signaling initiated by after wounding, the label-retaining cells are located
growth factors (109, 163). During tissue repair, at the subepithelial connective tissue underlying the
fibroblasts are exposed to a variety of extracellular wound edge and at the perivascular location. It
matrix molecules and soluble growth factors that can seems, however, that only a subpopulation of the lo-
exert multiple signals at the same time. Additionally, cal fibroblasts proliferate in response to tissue injury
cells experience tensional forces from the extracellu- (225), suggesting that certain fibroblast subsets are
lar matrix during cell migration and reorganization more responsive to growth stimulation than others.
of the wound matrix, which can modulate signaling Local fibroblasts are not the only source of wound
inside the cell (27, 205, 228). The function of extra- fibroblasts. After wounding, pericytes that are resi-
cellular matrix proteins may be further modulated dents of the surroundings of the vascular endo-
by proteolytic modification (13, 143, 227), and by thelium of capillaries and venules are also induced
conformational changes induced by tensional forces to proliferate and migrate into the wound (79, 106,
(295). Therefore, the regulation of fibroblast gene ex- 158). Although fibroblasts have multiple functions in
pression upon wounding is likely to be very complex wound repair, their hallmark characteristic is the
and dependent on the balance between different ac- property to produce collagen and other structural
tivated signaling pathways (41). extracellular matrix proteins that replace the blood
clot. This property separates wound fibroblasts from
inflammatory and vascular endothelial cells that are
Origin of wound fibroblasts
also involved in the formation of granulation tissue.
The multiple functions of wound fibroblasts raise Accordingly, upon wounding, the pattern of gene ex-
the question of whether all of the tasks are per- pression in pericytes is reprogrammed from a con-
formed by a single cell type or multiple different tractile to a migratory collagen-producing cell, sug-
phenotypes are involved (Table 2). If the latter case gesting that these cells are able to function as ma-
is correct, does the heterogeneity result from mi- trix-producing wound fibroblasts.
gration of phenotypically different cells into wound Could some of the wound fibroblasts also arise
or does it result from differentiation of the fibro- from bone marrow stem cells? Interestingly, early
blasts that have populated the wound? These ques- work by Cohnheim in 1867 suggested that some of
tions are still largely unanswered. There is evidence, the wound fibroblasts may originate from bone mar-
however, that connective tissue fibroblasts are het- row. This was further supported by recent findings
erogeneous in several properties, including, respon- showing that genetically marked bone marrow
siveness to growth factors and in the ability to pro- stromal cells serve as a source of progenitor cells for
duce specific extracellular matrix proteins (93, 94, various mesenchymal tissues. Most importantly,

Table 2. Origin of wound fibroblasts


Possible sources of wound fibroblasts Steps involved
Surrounding connective tissue Migration, differentiation
Pericytes Proliferation, migration
Bone marrow Systemic control, homing, differentiation

135
Häkkinen et al.

these cells apparently translocate to the target tissue able that the decision of the fibroblast to enter the
where they acquire the phenotype of the resident cell cycle depends on the repertoire of integrins that
cells (183, 193). These ‘‘peripheral blood fibrocytes’’ actively interact with specific extracellular matrix
accumulate in the wound already in the early acute proteins and on the combination of growth factors
phase but are also present in the scar tissue (15). that the cells are exposed to at the particular phase
Fibrocytes function as antigen presenting cells and of wound repair.
can prime naive T cells (25) and secrete a number of Cell migration is needed for fibroblasts to enter
cytokines involved in immune response, hematopo- the wound provisional matrix. Fibroblast migration
esis and extracellular matrix synthesis (25, 26), sug- into the blood clot occurs only after a lag period of
gesting that fibrocytes may play a role in the regula- 3 to 5 days, the time required for cells to proliferate
tion of the acute inflammatory reaction as well as in (32, 157). The lag phase may also be needed for the
the matrix deposition in wounds. cells to be activated from quiescent state to be fully
Taken together, the matrix-producing cells that active migratory cells and to be recruited from their
populate wound granulation tissue originate from original location in the tissue or bone marrow to the
diverse tissue sources and, depending on their ori- wound site. It is believed that, in order to detach
gin, they may serve different functions. from a collagen-rich matrix, fibroblasts have to
downregulate their expression of collagen receptors
and upregulate integrins that bind fibronectin, fi-
Integrin expression and function in fibroblasts brin, fibrinogen and vitronectin in the provisional
during wound repair
wound matrix. Accordingly, about 3 to 5 days after
In the connective tissue, fibroblasts are surrounded injury, fibroblasts that migrate into the blood clot ex-
by a matrix that contains collagen and cellular press the primary fibronectin receptor a5b1 integrin
fibronectin as the major components. Consequently, and a3b1 integrin while they show downregulation
quiescent fibroblasts express collagen receptors of collagen-binding a1 and a2 integrin expression at
a1b1 and a2b1 and the major fibronectin receptor the wound margin (74, 281). Platelet-derived growth
a5b1 integrin which they use for adhesion to the ma- factor, which is abundantly expressed during early
trix (74, 271, 281). Fibroblasts express a3b1 (281) and wound repair, stimulates cell migration towards
integrin heterodimers of the av subfamily (Häkkinen fibronectin by increasing the synthesis of a5 and a3
et al., unpublished) that can also be used to bind integrins (74, 117). On the other hand, platelet-de-
fibronectin (4). Although cultured fibroblasts are rived growth factor downregulates a1 (74) while it
able to express avb1, avb3 and avb5 integrins (70, stimulates a2 integrin expression by fibroblasts in
73, 92), it is not completely clear with which b sub- culture. Therefore, the regulation of a2 integrin ex-
units av actually combines to form integrin hetero- pression in vivo and in vitro is different, possibly be-
dimers in quiescent fibroblasts in vivo. In cell cul- cause of the multiple signals in addition to platelet-
ture, fibroblasts also express a4b1 integrin that binds derived growth factor that modulate integrin ex-
to fibronectin (72), but it is unclear whether this pression in the tissue. One of the factors that modu-
integrin is actually expressed by fibroblasts in vivo. lates integrin expression during wound repair is the
The b1 integrins in quiescent fibroblasts are par- composition of the extracellular matrix. If fibroblasts
tially in an inactive state and become activated after are cultured in fibrin and fibronectin-rich matrices
wounding in fibroblasts that are located near the de- that resemble the early wound matrix, they show
fect (Häkkinen et al., unpublished). Several lines of selective upregulation of a3 and a5 integrin mRNA
evidence show that integrin activation and integrin- as opposed to cells that are in collagen-rich matrix,
mediated cell adhesion to extracellular matrix is im- which show upregulation of a2 integrin (281). This
portant for induction of DNA synthesis (179). Speci- suggests that signals initiated by growth factors and
fic integrin-ligand binding is required to commence extracellular matrix molecules collaborate to induce
DNA synthesis of cells. Cell adhesion to extracellular appropriate integrin expression to support cell mi-
matrix via integrins strongly influences the ability of gration on fibrin-fibronectin matrix in the early
normal cells to respond to soluble mitogens (43, 76, wound repair and to allow cell adhesion on collagen
109, 221). Activated integrins can physically complex later during scar formation.
with growth factor receptors (see above) which pro- In the early wound, fibroblast migration seems to
vides a mechanism of how different signaling mol- be primarily mediated by fibronectin because
ecules may be targeted to cell-matrix interaction fibroblast migration from a three-dimensional colla-
sites inside the cell (161, 218). It is, therefore, prob- gen matrix into a fibrin clot depends on the presence

136
Cell biology of gingival wound healing

of fibronectin in the matrix and can be blocked with for several cell surface receptors, including avb3
antibodies against a5b1 and avb3 integrins in vitro integrin in fibroblasts that binds to the RGD se-
(84). Fibroblasts can also use avb3 and avb5 inte- quence found in the third fibronectin type repeat of
grins for cell adhesion on vitronectin present in the the tenascin molecule. Ligand binding by integrins
clot (70) and avb3 for cell adhesion and migration to this repeat is modulated by other counteradhesive
on fibrin and fibrinogen, respectively (71, 84). Cell domains in the molecule (40).
migration is regulated by the structural organization Thrombospondin can also, like tenascin-C, modu-
of the fibronectin matrix. In the normal connective late cell adhesion, migration and proliferation (83).
tissue, cells deposit and arrange fibronectin into a It contains an RGD sequence that is recognized by
fibrillar network between the cells and other matrix avb3 integrin expressed by fibroblasts (240, 255), but
molecules using primarily a5b1 and possibly avb3 it also contains, like tenascin-C, additional binding
integrins (279, 280, 287). In contrast, in the blood sites for other cell surface receptors (240). SPARC is
clot, plasma fibronectin is cross-linked with fibrin to also able to interfere with integrin-mediated cell ad-
form a three-dimensional scaffold (224). The vari- hesion on extracellular matrix, and this is believed
ation in the architecture of the fibronectin network to occur via interaction with other cell surface mol-
can regulate multiple cell functions, suggesting that ecules that are able to bind SPARC and not with
fibronectin in the connective tissue and in the blood steric disruption of integrin–extracellular matrix in-
clot may elicit different signals (224, 233). During teraction (168). Because of their different temporal
wound repair, cells are induced to deposit alterna- expression pattern during wound repair, it is poss-
tively spliced isoforms of cellular fibronectin con- ible that tenascin-C, thrompospondin and SPARC
taining either the EIIIA or EIIIB modules (see above). regulate different cellular functions. Indeed, gene
The EIIIA or EIIIB modules have different properties knockout animal studies have indicated that throm-
to support cell adhesion, migration, and prolifer- bospondin plays a role in regulation of the activity
ation (95, 150, 151), suggesting that they may have of transforming growth factor-b, collagen fiber or-
different functions in wound repair. ganization and vascularization in the connective
Wounding induces deposition of extracellular ma- tissue (39, 126), which are all important processes in
trix proteins that modulate cell adhesion. Among the physiology of wound repair.
these molecules, tenascin-C, thrombospondin and
secreted protein, rich in cysteine/osteonectin
Wound contraction
(SPARC) are multifunctional molecules that can
serve as ligands for multiple cell surface receptors One of the tasks of fibroblasts in granulation tissue
(83). Typically, the expression of tenascin-C and is to bring the wound margins closer together to
thrombospondin is potently stimulated early during allow rapid wound closure. It is not completely clear
granulation tissue formation, while SPARC is ex- how wound contraction actually happens in vivo.
pressed from middle to late stages of repair. Express- The most widely held concepts suggest that wound
ion of tenascin-C and SPARC remains elevated dur- closure follows when central granulation tissue is
ing tissue reorganization while the expression of contracted either by myofibroblasts pulling the new
thrombospondin is only transient (50, 144). Interest- collagen matrix (69) and/or by fibroblasts changing
ingly, wounding specifically induces the expression the organization of the wound matrix by migrating
of the large tenascin-C splice isoform (144, Häkkinen through it (54, 67). On the other hand, experimental
et al., unpublished) that is normally expressed pre- removal of central granulation tissue does not seem
dominantly during embryonic development (156, to prevent wound contraction, suggesting an alter-
192). Tenascin-C contains both adhesive and native mechanism in which polarized coordinated
counteradhesive domains, although mostly it is be- migration of a rim of densely packed fibroblasts pull
lieved to reduce the adhesion of cells to fibronectin forward the wound edges (89).
and therefore enhance cell migration (253). The ef- Myofibroblasts are likely to play a role in wound
fect of tenascin-C on cell adhesion to fibronectin can contraction and matrix deposition. This is supported
also further modulate gene expression by the cells by findings that myofibroblasts are typically present
(253). Interestingly, the large tenascin-C splice iso- in tissues that are under mechanical stress but they
form functions differently in regulation of cell ad- are only occasionally found in normal tissue (46).
hesion and in binding fibronectin as compared with Myofibroblasts become especially prominent in
the more ubiquitously expressed small isoform (29, pathological conditions including wound repair,
63, 64, 113, 169, 184). Tenascin-C can serve as ligand tissue fibrosis and tumor stroma (42, 46, 201). These

137
Häkkinen et al.

cells are characterized by presence of abundant myofibroblasts disappear because of apoptosis and
cytoplasmic microfilament bundles and a-smooth the scar becomes less cellular and new fibroblasts
muscle actin. They differ from normal fibroblasts with properties typical to normal connective tissue
morphologically, in growth potential, expression of fibroblasts emerge (42, 48, 217). Apoptosis of myo-
proteoglycans and collagen, responsiveness to trans- fibroblasts begins at day 12, peaks at day 20 and re-
forming growth factor b, organization of focal ad- solves by day 60 after wounding (48). Factors that in-
hesions, and in their ability to organize cellular hibit a-smooth muscle actin expression and differen-
fibronectin in culture (51, 93, 94, 212). Fibroblasts tiation of myofibroblasts are not well characterized. It
cultured from wound granulation tissue are rich in has been noted that interferon-g has an antifibrotic
myofibroblasts and express a similar repertoire of effect, which probably results from its ability to inhibit
integrins as normal connective tissue fibroblasts. fibroblast differentiation into myofibroblasts (217).
They are, however, phenotypically unique in that Additionally, myofibroblasts are more sensitive to
they show reduced cell spreading on fibronectin as basic fibroblast growth factor–induced apoptosis
a result of altered interaction of a5b1 integrin and than normal connective tissue fibroblasts (68), which
fibronectin (92). may provide a mechanism to reduce the amount of
The differentiation of myofibroblasts occurs be- myofibroblasts in the tissue when they are no longer
tween 6 and 15 days after wounding (42, 217). After needed. Interestingly, myofibroblasts persist in cer-
15 days, about 70% of fibroblasts in granulation tain pathological conditions, including hyperthroph-
tissue express a-smooth muscle actin. Differen- ic and fibrotic lesions of many organs, suggesting that
tiation of myofibroblasts coincides with wound con- they are involved in the accumulation of extracellular
traction, which has led to the idea that these cells matrix that is characteristic for these lesions (45, 217).
are actually involved in this process. The differen- Therefore, for normal wound repair it is important
tiation of myofibroblasts is induced by transforming that the myofibroblasts undergo apoptosis and are re-
growth factor b in vivo and in cell culture but not placed by normal fibroblasts when wound contrac-
with other profibrotic growth factors and cytokines tion is completed.
(47, 204). Importantly, myofibroblasts differentiation An interesting question is where the fibroblasts that
is controlled by integrin mediated mechanisms and replace myofibroblasts and that eventually maintain
depends on the composition of the extracellular ma- the connective tissue structure during tissue homeo-
trix and the tensional forces mediated from the ma- stasis originate from. Because these cells have pheno-
trix. Recent studies have shown that the induction of typic properties characteristic to normal connective
myofibroblast differentiation by transforming tissue fibroblasts, it is possible that they are derived
growth factor-b requires deposition of cellular EIIIA from the connective tissue next to the wound or, alter-
fibronectin in the pericellular matrix (226). In tissue, natively, they differentiate from more primitive
differentiation of wound fibroblasts into myofibro- fibroblasts that initially populate the wound. Regard-
blasts follows induction of EIIIA fibronectin express- less of their origin, it seems that replacement of the
ion (226). The induction of the expression of a- wound fibroblasts with cells of normal connective
smooth muscle actin in fibroblasts by transforming tissue fibroblast phenotype is necessary for the com-
growth factor b is also regulated by the changes in plete regeneration of the tissue.
the collagen matrix around the cells and on the de-
velopment of intracellular tension (5, 171). This is in
Mechanical tension
turn regulated by interaction between a2b1 integrins
and collagen (171). The induction of a-smooth The interactions between cells and extracellular ma-
muscle actin expression during wound repair is trix during granulation tissue formation can be
thought to provide a mechanism to stop fibroblast studied in cell culture using cell-populated three-di-
migration when cells reach their destination. Ter- mensional fibrin or collagen gels (86, 256). In prin-
mination of migration and formation of prominent ciple, the fibrin gels correspond to the situation in
focal adhesions may be needed to promote stable early wound repair when fibroblasts have migrated
(200) cell adhesion to matrix, which is required for into the fibrin containing provisional matrix while
myofibroblasts to acquire a matrix-synthesizing and the collagen gel model resembles later phase of
contractile phenotype. wound repair when granulation tissue fibroblasts re-
In ideal wound repair, scar tissue is remodeled and side in a newly deposited collagen-rich matrix that
reorganized to form structurally and functionally nor- undergoes remodeling. In these models, fibroblasts
mal connective tissue. When contraction stops, the attach to the proximal fibrin or collagen fibers and

138
Cell biology of gingival wound healing

in an attempt to migrate pull the fibers together to and mediate signals that promote cell growth (269,
form a dense tissue-like structure. When fibroblasts 294), differences in fibronectin receptor engagement
are allowed to grow in fibrin gels they eventually re- may be a mechanism of how cell proliferation is
place the fibrin matrix with newly deposited colla- regulated by mechanical signals. Therefore, reduc-
gen (256). If the gel is not relaxed, fibroblasts trying tion of mechanical strain during tissue maturation
to pull the fibers experience high tension from the may provide a mechanism to downregulate cell
matrix that resembles wound granulation tissue. growth and induce apoptosis to normalize the cellu-
Contraction of three-dimensional collagen gel is me- larity in the tissue. Additionally, in the later phases
diated by integrins and can be stimulated by serum of wound repair the activity of growth factors is
and growth factors present in wound, including lyso- gradually downregulated (231, 288), which will
phosphatidic acid, transforming growth factor b and further reduce the number of growth stimulatory sig-
platelet-derived growth factor (34, 87, 90, 164, 197, nals for fibroblasts. In the regulation of cell growth
250). Currently, there is some controversy over which and apoptosis in the granulation tissue, integrins
specific integrin-ligand interactions actually mediate probably play an important role because they me-
collagen contraction. Binding between fibroblasts diate the signals initiated by mechanical tension and
and collagen fibrils is evidently the first and most collaborate with growth factor signaling and mediate
important step in the collagen gel contraction. Most signals that regulate cell survival (109, 205, 206, 228).
data show that in human connective tissue fibro- Interestingly, fibroblasts undergo apoptosis in con-
blasts, collagen gel contraction is mediated by the tractile collagen gels but not in fibrin gels (66), sug-
collagen receptor a2b1 integrin (214, 238). The rate gesting that specific types of integrin–extracellular
of contraction depends on the amount of a2b1 inte- matrix interactions are needed for initiation of
grins expressed by the cells (197). This is in accord- apoptosis. This may provide a mechanism to prevent
ance with simultaneous induction of a2 integrin ex- fibroblasts from undergoing apoptosis in the early
pression by wound fibroblasts with formation of col- fibrin-rich provisional wound matrix and to allow
lagenous scar and beginning of wound contraction their proliferation, which is needed to populate the
in vivo (282). granulation tissue with fibroblasts.
An additional mechanism that is likely to regulate
cell growth in the granulation tissue is the structural
Role of integrins in regulation of cell proliferation organization of the extracellular matrix. During
and survival in granulation tissue
granulation tissue formation, fibroblasts first syn-
During the formation of granulation tissue, fibro- thesize fibronectin and then type I collagen that is
blasts that emerge into the provisional wound matrix gradually organized from monomeric molecules to
stop migrating and undergo cell proliferation. Ac- form a fibrillar collagen matrix (125). Monomeric
cordingly, about 7 to 10 days after wounding, the collagen through a2b1 integrin promotes cell pro-
granulation tissue becomes hypercellular containing liferation while fibrillar collagen downregulates it
abundant fibroblasts and endothelial cells (157, 271). (123). Additionally, studies using fibronectin de-
After that, during tissue maturation, the cell number ficient cells have shown that assembly of fibronectin
in the granulation tissue gradually decreases (48, matrix by the cells is required for fibronectin to pro-
157, 271). It appears that the organization of collagen mote cell growth on different extracellular matrix
and mechanical tension mediated from the extracel- molecules (233). Therefore, it is probable that the de-
lular matrix are important factors that modulate position and organization of extracellular matrix by
DNA synthesis and cell survival. This is evidenced by fibroblasts provides a feedback mechanism to regu-
the finding that fibroblasts cultured under mechan- late cell growth during wound repair.
ical tension proliferate actively, but after stress relax-
ation the DNA synthesis is rapidly downregulated
and cells start to undergo apoptosis (66, 87, 139, Regulation of protein synthesis and matrix
degradation in the granulation tissue
162). Stress relaxation also downregulates autophos-
phorylation of growth factor receptors, which con- Remodeling of tissue during wound repair requires
tributes to the growth reduction (139, 159, 241). In a controlled synthesis and degradation of extracellular
mechanically stressed collagen gel, cells develop an matrix. The most important factors that regulate
organized cell surface fibronectin network (162, 251) synthesis and secretion of these molecules include
that disappears after stress relaxation. Since a5b1 growth factors and signals from the extracellular ma-
integrin can probably serve as a mechanoreceptor trix. There is evidence that integrin binding to its

139
Häkkinen et al.

ligand can directly induce gene expression (102, 254, teinase-13 expression occurs by a2b1 and by both
272). Most importantly, there seems to be a specific a1b1 and a2b1 integrins, respectively (195, 197).
integrin-ligand interaction that is needed for stimu- Integrin a1b1 seems to be able to function as a feed-
lation of specific type of protein synthesis (254, 272). back regulator of collagen synthesis in fibroblasts
In this process, the signals mediated by integrins are (75).
further modulated by growth factors, the structural
architecture of the extracellular matrix as well as by
tensional forces (109, 202, 228). Gingival wound repair: similarities to scarless fetal
wound repair
During granulation tissue formation, fibroblasts
are stimulated by transforming growth factor-b to There has been a general observation that wounds
deposit new extracellular matrix proteins. Interest- in the oral mucosa heal faster and with less scarring
ingly, early matrix deposition seems to occur first at than extraoral wounds (Table 3). Scar tissue is char-
about day 7 after injury in the granulation tissue im- acterized by excessive accumulation of disorderly ar-
mediately under the newly formed epithelium (178). ranged collagen, mostly type I and III (219), proteo-
This coincides with the peak in activation of latent glycans (274, 289) and persistent myofibroblasts (42,
transforming growth factor-b from storages in the 46, 201), which leads to aberrant function of the
blood clot provisonal matrix and in the matrix of the tissue. Surprisingly, there are only a few reports that
granulation tissue (81, 288) and with induction of have quantitatively tested this hypothesis. Neverthe-
epithelial avb6 integrin which may regulate the acti- less, based mostly on animal studies it seems that
vation of latent transforming growth factor-b (see wound healing in oral mucosa, indeed, is faster and
above, 91, Häkkinen et al., unpublished). Once acti- results in less scarring than in skin (223, 286). It is
vated, transforming growth factor-b can induce its probable that oral wound healing is enhanced partly
own production by fibroblasts (139), which may be a because of factors present in the saliva and by the
mechanism to maintain high activity of transforming specific microflora of the oral cavity (see below). Ad-
growth factor-b in the granulation tissue. ditionally, the properties of the cells involved in
As discussed earlier, a potent modulator of protein tissue regeneration in oral mucosa are unique and
synthesis is mechanical tension. Generally, when share properties of fetal cells (219). Scar formation is
cells are grown in collagen gels under tension they developmentally regulated because, in early ges-
show a relatively high protein synthesis rate, while tation, fetal wound repair occurs without scar forma-
after stress relaxation the protein synthesis is down- tion and the transition to healing with scar forma-
regulated (162). The signals initiated by tension in tion occurs late in the gestation (56, 104). Interest-
a three-dimensional collagen matrix require specific ingly, the initiation of scar formation parallels the
integrin-collagen interactions and may lead to speci- induction of myofibroblast differentiation during
fic gene activation. Additionally, platelet-derived wound repair at different stages of embryonal devel-
growth factor–induced upregulation of a2 integrin opment (56), suggesting that phenotypic modulation
expression is stimulated if fibroblasts are cultured in of wound fibroblasts into myofibroblasts may be in-
collagen gel while expression of a5 and a3 integrins volved in scar formation.
is attenuated (281). Transforming growth factor-b As compared with adult wounds, fetal wounds are
can also potently stimulate expression of a2 integrin characterized by the absence of clot formation and
in fibroblasts that interact with collagen, and this de- inflammatory reaction. They also show unique spa-
pends on the tensional forces that the cells experi- tial and temporal organization of extracellular matrix
ence (5). It seems that tension mediated from the and reduced expression of transforming growth fac-
matrix can effectively regulate the balance between tor-b (274). Several investigations have compared the
matrix production and degradation. For example, properties of adult skin and gingival fibroblasts with
upon stress relaxation the expression of type I colla- fetal fibroblasts. The findings indicate that, unlike
gen is downregulated while that of matrix metallo- adult skin fibroblasts, adult gingival fibroblasts
proteinase-1 and -13 is upregulated (30, 195, 197, located in the papillary connective tissue share
252). The regulation of collagen and matrix metallo- many properties with fetal fibroblasts, including
proteinase genes by collagen matrix is partially regu- their growth and migration properties, cell mor-
lated by different collagen receptors. The downregu- phology, production of migration stimulating factor,
lation of collagen synthesis after stress relaxation is and production and response to cytokines (219). Ad-
mediated by a1b1 integrin and the upregulation of ditionally, similar to fetal fibroblasts, oral mucosal or
matrix metalloproteinase-1 and matrix metallopro- gingival fibroblasts are able to contract three-dimen-

140
Cell biology of gingival wound healing

Table 3. Special features of oral wound healing


Factor Mechanism
Saliva Moisture, ionic strength
Growth factors (EGF, TGFb, FGF, IGF etc.)
Unknown factors
Bacteria Stimulation of macrophage influx
Direct stimulative action on keratinocytes and fibroblasts
Phenotype of cells Fetal-like fibroblasts with unique response
Specialized epithelium and connective tissue

sional collagen matrix faster as dermal fibroblasts healing of oral wounds in people with xerostomia
(105, 237). They also populate experimental wounds or sialadenectomized animals (12, 55, 103). Animals
faster than their dermal counterparts in culture (2). instinctly lick their wounds, which appears to result
Gingival fibroblasts also differ from dermal fibro- in faster healing (11). There are several physico-
blasts in their ability to secrete proteolytic enzymes. chemical factors in saliva favoring gingival healing.
Matrix metalloproteinase-13, a potent collagenase These include an appropriate pH, ionic strength, and
(273), is expressed in the granulation tissue during presence of ions such as calcium and magnesium
acute wound repair at 7- and 14-day-old human gin- required for healing (53). Saliva also has an efficient
gival wounds but not in dermal wounds (296). Ad- capacity to reduce redox activity caused by tran-
ditionally, when gingival fibroblasts are inoculated sitional metal ions and inhibit the production of free
into three-dimensional fibrin matrix they are able to radicals that may be beneficial for the healing pro-
reorganize and degrade the matrix rapidly. This is cess (170). Lubrication of oral mucosa provided by
because of high expression of tissue plasminogen ac- saliva is beneficial for wound healing. The advan-
tivator. Matrix reorganization and fibrinolysis is less tageous effects of maintaining a moist wound en-
advanced in dermal fibroblast-inoculated matrices vironment include prevention of tissue dehydration
(141, 142). The regulation of fibrinolysis by gingival and cell death, accelerated angiogenesis and in-
fibroblasts depends on the tensional forces that the creased breakdown of fibrin and tissue debris.
cells experience from the matrix (140). The various Hence, the use of hydrocolloid occlusive dressing is
findings described above suggest that several cell a useful adjunct in facilitating cutaneous wound
functions important in tissue repair are shared by healing (61). The therapeutic effect of saliva on heal-
fetal and gingival fibroblasts and differ from dermal ing of skin wounds has been demonstrated exper-
fibroblasts. It is possible that gingival fibroblasts are imentally in calves. Compared to saline-treated
phenotypically unique cells in adult tissue that may wounds, saliva-treated wounds had shorter in-
contribute to the rapid healing of oral wounds with flammatory reaction and faster epithelial coverage
minimal scarring in the gingiva. and connective tissue regeneration (268). It appears
that moisture and ionic strength are not the primary
factors in saliva that promote tissue repair. This po-
tential is probably due to the presence of several fac-
Role of saliva and gingival tors in saliva including various growth factors and
crevicular fluid in oral wound bacteria (293).
healing Growth factors found in saliva are synthesized by
salivary glands or derived from plasma through gin-
While it is clear that the excellent healing potential gival crevice. Epithelial cells and connective tissue
of oral mucosa results to a large extent from the in- cells also produce their own growth factors that act
trinsic tissue factors such as the presence of struc- either in a paracrine or autocrine manner. Because
tural cells with potential for tissue regeneration, many of the growth factors are transported to saliva
dense vasculature and high turnover rate of connec- along with gingival crevicular fluid, it is conceivable
tive tissue and epithelium, it is also apparent that that their concentrations in gingival tissue are higher
saliva provides a unique environment in the mouth than elsewhere in the oral cavity. Therefore the peri-
conducive for rapid tissue repair (Table 3). This be- odontium is in a favorable position with respect to
comes obvious from the studies showing delayed tissue healing.

141
Häkkinen et al.

As discussed above, wound healing is a complex of inflammation and tissue destruction or increased
phenomenon involving increased proliferation, ad- cell growth and tissue repair.
hesion and migration of cells of connective tissue Integrin ligands such as collagen, vitronectin,
and epithelium, inflammatory reactions and re- fibronectins and laminins are involved in directing
modeling of extracellular matrix. All these phenom- the adhesion and migration of cells (261). It is there-
ena are directed by growth factors (9, 10, 82). Differ- fore possible that these molecules, when present in
ent growth factors have specific functions and target gingival crevicular fluid or saliva, may modulate
cells in wound healing, and their delicate balance is wound healing of gingiva. Indeed, fibronectin has
required for optimal tissue repair. The actual roles of been found both in crevicular fluid and saliva (112,
each growth factor in saliva are not known. The best 129, 257). Even though not yet reported, it is reason-
studied salivary growth factor, and possibly the most able to assume that other adhesion molecules may
important one, is epidermal growth factor. In his be present in saliva.
pioneer studies, Cohen found that a protein compo- Extracellular matrix remodeling is an essential
nent of mouse submandibular gland induced pre- part of wound repair. In this process matrix metallo-
cocious eyelid opening and incisor tooth eruption. proteinases and other neutral proteinases have an
Later it was discovered that the factor termed epider- important function. During periodontal diseases
mal growth factor has a multitude of effects on cell gingival crevicular fluid and saliva are rich in col-
proliferation, cell migration and extracellular matrix lagenases, gelatinases and elastases (133–136, 149,
metabolism (21, 37, 220). It has now become obvious 232, 263–265). Most of these enzymes appear to de-
that epidermal growth factor is needed for the nor- rive from neutrophils migrating from inflamed peri-
mal maintenance and repair of oral mucosa (174). odontium. However, salivary gland cells and acti-
Interestingly, salivary epidermal growth factor also vated epithelial cells and fibroblasts of periodontium
plays a role in the maintenance of gastric and ileal produce their own matrix metalloproteases includ-
mucosal integrity (194, 213). In humans and many ing collagenase-1 (matrix metalloprotease-1), col-
other animals, salivary glands are the major epider- lagenase-3 (matrix metalloprotease-13) and gela-
mal growth factor–producing organs. In humans epi- tinase A (matrix metalloprotease-2) (209, 211, 258).
dermal growth factor is synthesized by both parotid In concert the matrix metalloproteases and elastases
and submandibular glands. During oral wound heal- are capable of cleaving all extracellular matrix pro-
ing, the concentration of epidermal growth factor is teins and proteoglycans. As discussed earlier, proteo-
increased in saliva (99, 180, 181). lytic enzymes are necessary for proper wound heal-
Another growth factor found in saliva is the vascu- ing. These matrix metalloproteases could also re-
lar endothelial growth factor (243). This protein is lease cryptic bioactive domains from matrix
important in many aspects of angiogenesis and in- molecules that may regulate cell proliferation and
flammation such as endothelial growth, permeability migration (285). For example, fibronectin and colla-
and leukocyte adherence (52). A number of other gen fragments have been demonstrated in gingival
growth factors are also present in saliva. These in- crevicular fluid (245) and saliva (77). Practically no
clude nerve growth factor and members of trans- information presently exists on the possible role of
forming growth factor b, fibroblast growth factor and these extracellular matrix fragments in promoting
insulin-like growth factor families (230). As these healing of oral wounds.
substances have specific regulatory roles in cell
growth and extracellular matrix formation, they are
Role of bacteria in oral wound healing
important in maintaining health of the oral cavity
and in healing of oral mucosal tissues (293). The oral cavity harbors considerable amounts of
The presence of growth factors in crevicular fluid bacteria. More than 500 bacterial species have been
has received limited attention. The major focus of so far identified in the oral cavity (165). Recognizing
the studies has been pro-inflammatory cytokines, the limitation of the present detection methods, in
such as interleukin-1 and tumor necrosis factor a reality several times more species may colonize in
(177). It is important to recognize that while one ac- the mouth. It is clear that bacteria affect wound
tion of the crevicular fluid cytokines is promotion of healing in the oral cavity, and it is well established
inflammation, another function is to facilitate tissue that wounds colonized by pathogenic bacteria have
repair. Maintenance of the normal tissue turnover delayed healing (198, 249). Clinicians are aware of
results from a delicate balance of the cytokines. An the painful complications in extraction wound repair
alteration in this balance results either in persistence that result from bacterial infection (38, 57). Much

142
Cell biology of gingival wound healing

less attention has been given to the fact that small could modulate oral wound healing. For example, A.
concentrations of bacteria may increase the rate of actinomycetemcomitans GroEL-like heat-shock pro-
wound healing. tein and phospholipase C are able to modulate epi-
In 1921, Carrel reported that wounds of dogs thelial cell growth and cell migration (62, 80, 294).
treated with certain concentrations of Staphylococ- Specific mechanisms of these effects remain to be
cus aureus healed faster than untreated wounds. explored.
Since then several studies have confirmed that find-
ing, also using other bacterial species. Many factors
may contribute to this effect. Inflammatory reaction Conclusion
that is a prerequisite for tissue repair is accentuated
by bacterial contamination. Bacteria present in a Wound healing in the oral cavity is a very complex
wound will attract macrophages into the area and process. We are just starting to uncover the complex
induce their cytokine secretion. As a consequence, interplay between various cell types, growth factors
blood supply and granulation tissue formation are and salivary components. The focus of this chapter
accentuated in the wound. Proliferation of mes- was to summarize the two major events of gingival
enchymal cells is increased and synthesis rate of wound healing, namely re-epithelialization and the
connective tissue components is stimulated, leading formation of granulation tissue. Because of the
to greater tensile strength of the contaminated unique environment of oral cavity, we also reviewed
wounds in the course of healing (115, 127, 128, 136). the special features of oral healing. The interplay be-
Bacteria contain a variety of substances, some tween oral cells and their extracellular matrix, bac-
stimulating host cell proliferation and others teria, saliva and gingival crevicular fluid involves
exerting toxic effects. In addition, the same sub- myriad factors dictating the nature of the tissue re-
stance can be either stimulatory or inhibitory, de- pair process. It would be an enormous if not imposs-
pending on its concentration in tissue. Bacteria may ible task to sort out all these factors. Some of the
act directly on epithelial cells and connective tissue factors are already relatively well understood and
cells in wounds and, depending on the type and could be used for practical applications. Active
concentration, either accelerate or delay wound re- studies on certain growth factors are underway in
generation. We found that proliferation of gingival order to provide new tools for periodontal therapy
fibroblats in culture was increased by Prevotella in- (101). Similar studies utilizing other factors benefi-
termedius but decreased by the same concentrations cial for wound healing can be expected when more
of Porphyromonas gingivalis (133). Interestingly, basic research has been done elucidating their speci-
there was a great variation in this effect between fic functions. Only then we can design precision
fibroblast populations obtained from different pa- tools to speed-up (or slow-down) re-epithelializ-
tients. These findings imply that the potential for ation, granulation tissue formation and scarless
periodontal repair depends both on the bacterial wound healing. Modulation of cell–to–extracellular
flora and the individual cell populations of the peri- matrix adhesion will be the key target for computer
odontal wounds. Some bacterial factors have direct designed drugs engineered to guide optimal tissue
fibroblast and epithelial cell stimulating properties. repair.
Lipopolysaccharide of both Actinobacillus actino-
mycetemcomitans and P. gingivalis slightly increases
cell growth in vitro (98, 189). At higher concen- Acknowledgments
trations lipopolysaccharide from different bacterial
species inhibits cell proliferation (8, 134). Lipopoly- We thank Colin Wiebe for critical reading of the
saccharide as well as bacterial plaque extracts in- manuscript. Wound-healing studies performed in
crease hyaluronan production in cultured gingival the laboratories of the authors are supported by the
fibroblasts (7, 130). Hyaluronan is a high-molecular- Medical Research Council of Canada.
weight polyanionic glycosaminoglycan that plays an
important role in wound healing through specific in-
teraction with other matrix molecules and cells (23). References
S. aureus lipoteichoic acid and protein A induce
fibroblast production of hepatocyte growth factor, 1. Akiyama SK, Yamada SS, Chen W-T, Yamada KM. Analysis
which stimulates epithelial proliferation (6). There of fibronectin receptor function with monoclonal anti-
are numerous other factors present in bacteria that bodies: roles in cell adhesion, migration, matrix assembly,

143
Häkkinen et al.

and cytoskeletal organization. J Cell Biol 1989: 109: 863– 20. Cavani A, Zambruno G, Marconi A, Manca V, Marchetti M,
875. Giannetti A. Distinctive integrin expression in the newly
2. al-Khateeb T, Stephens P, Shepherd JP, Thomas DW. An forming epidermis during wound healing in humans. J
investigation of preferential fibroblast wound repopu- Invest Dermatol 1993: 101: 600–604.
lation using a novel in vitro wound model. J Periodontol 21. Chen JD, Kim JP, Zhang K, Sarret Y, Wynn KC, Kramer RH,
1997: 68: 1063–1069. Woodley DT. Epidermal growth factor (EGF) promotes
3. Anatoniades HN, Galanopoulos T, Neville-Golden J, Kirit- human keratinocyte locomotion on collagen by increas-
sy CP, Lynch E. Injury induces in vivo expression of plate- ing the a2 integrin subunit. Exp Cell Res 1993: 209: 216–
let-derived growth factor (PDGF) and PDGF receptor 223.
mRNAs in skin epithelial cells and PDGF mRNA in con- 22. Chen JWC, Rogers AA, Lydon MJ. Characterization of bio-
nective tissue fibroblasts. Proc Natl Acad Sci U S A 1991: logic properties of wound fluid collection during early
88: 565–569. stages of wound healing. J Invest Dermatol 1992: 99: 559–
4. Aplin AE, Howe A, Alahari SK, Juliano RL. Signal transduc- 564.
tion and signal modulation by cell adhesion receptors: 23. Chen WY, Abatangelo G. Functions of hyaluronan in
the role of integrins, cadherins, immunoglobulin-cell ad- wound repair. Wound Repair Regen 1999: 7: 79–89.
hesion molecules, and selectins. Pharmacol Rev 1998: 50: 24. Cherny RC, Honan MA, Thiagaran P. Site-directed mutag-
197–263. enensis of the arginine-glycine-aspartic acid in vitro nec-
5. Arora PD, Narani N, McCulloch CA. The compliance of tin abolishes cell adhesion. J Biol Chem 1993: 268: 9725–
collagen gels regulates transforming growth factor-b in- 9729.
duction of a-smooth muscle actin in fibroblasts. Am J 25. Chesney J, Bacher M, Bender A, Bucala R. The peripheral
Pathol 1999: 154: 871–882. blood fibrocyte is a potent antigen-presenting cell cap-
6. Baroni A, Perfetto B, Ruocco E, Rossano F. Lipoteichoic able of priming naive T cells in situ. Proc Natl Acad Sci
acid and protein-A from Staphylococcus aureus stimulate U S A 1997: 94: 6307–6312.
release of hepatocyte growth factor (HGF) by human der- 26. Chesney J, Metz C, Stavitsky AB, Bacher M, Bucala RJ.
mal fibroblasts. Arch Dermatol Res 1998: 290: 211–214. Regulated production of type I collagen and inflamma-
7. Bartold PM. Lipopolysaccharide stimulation of hyaluron- tory cytokines by peripheral blood fibrocytes. Immu-
ate synthesis by human gingival fibroblasts in vitro. Arch nology 1998: 160: 419–425.
Oral Biol 1991: 36: 791–797. 27. Chiquet M, Matthisson M, Koch M, Tannheimer M,
8. Bartold PM, Narayanan AS, Page RC. Platelet-derived Chiquet-Ehrismann R. Regulation of extracellular matrix
growth factor reduces the inhibitory effects of lipopoly- synthesis by mechanical stress. Biochem Cell Biol 1996:
saccharide on gingival fibroblast proliferation. J Peri- 74: 737–744.
odontal Res 1992: 27: 499–505. 28. Chiquet-Ehrismann R. What distinguishes tenascin from
9. Bennett NT, Schultz GS. Growth factors and wound heal- fibronectin? FASEB J 1990: 4: 2598–2604.
ing: biochemical properties of growth factors and their 29. Chiquet-Ehrismann R, Matsuoka Y, Hofer U, Spring J,
receptors. Am J Surg 1993: 165: 728–737. Bernasconi C, Chiquet M. Tenascin variants: differential
10. Bennett NT, Schultz GS. Growth factors and wound heal- binding to fibronectin and distinct distribution in cell cul-
ing. II. Role in normal and chronic wound healing. Am J tures and tissues. Cell Regul 1991: 2: 927–938.
Surg 1993: 166: 74–81. 30. Chiquet-Ehrismann R, Tannheimer M, Koch M, Brunner
11. Bodner L. Effect of parotid submandibular and sublingual A, Spring J, Martin D, Baumgartner S, Chiquet M. Tenas-
saliva on wound healing in rats. Comp Biochem Physiol cin-C expression by fibroblasts is elevated in stressed col-
A 1991: 100: 887–890. lagen gels. J Cell Biol 1994: 127: 2093–2101.
12. Bodner L, Dayan D. Epithelium and connective tissue re- 31. Chuong CM, Chen HM. Enhanced expression of neural
generation during palatal wound healing in desalivated cell adhesion molecules and tenascin (Cytotactin) during
rats – a comparative study. Comp Biochem Physiol A Phy- wound healing. Am J Pathol 1991: 38: 427–440.
siol 1995: 111: 415–419. 32. Clark RA. Regulation of fibroplasia in cutaneous wound
13. Boudreau N, Bissell MJ. Extracellular matrix signalling: in- repair. Am J Med Sci 1993: 306: 42–48.
tegration of form and function in normal and malignant 33. Clark RA. Overview and general considerations. In: Clark
cells. Curr Opin Cell Biol 1998: 10: 640–646. RAF, ed. The molecular and cellular biology of wound re-
14. Bourdon MA, Ruoslahti E. Tenascin mediates cell attach- pair. New York: Plenum Press, 1996: 3–50.
ment through an RGD-dependent receptor. J Cell Biol 34. Clark RA, Folkvord JM, Hart CE, Murray MJ, McPherson
1989: 108: 1149–1155. JMJ. Platelet isoforms of platelet-derived growth factor
15. Bucala R, Spiegel LA, Chesney J, Hogan M, Cerami A. Cir- stimulate fibroblasts to contract collagen matrices. J Clin
culating fibrocytes define a new leukocyte subpopulation Invest 1989: 84: 1036–1040.
that mediates tissue repair. Mol Med 1994: 1: 71–81. 35. Clark RAF, Lanigan JM, DellaPelle P, Manseau E, Dvorak
16. Burke RD. Invertebrate integrins: structure, function, and HF, Colvin RB. Fibronectin and fibrin provide a pro-
evolution. Int Rev Cytol 1999: 191: 257–284. visional matrix for epidermal cell migration during
17. Busk M, Pytela R, Sheppard D. Characterization of the wound reepithelialization. J Invest Dermatol 1982: 70:
integrin avb6 as a fibronectin-binding protein. J Biol 264–269.
Chem 1992: 267: 5790–5796. 36. Clark RAF, Spencer J, Larjava H, Ferguson MWJ. Re-epi-
18. Carrel A. Cicatrization of wounds. XII. Factors initiating thelialization of normal human excisional wounds is as-
regeneration. J Exp Med 1921: 34: 425–432. sociated with a switch from avb5 to avb6 integrins. Br J
19. Carter WG, Ryan MC, Gahr PJ. Epiligrin, a new cell ad- Dermatol 1996: 135: 46–51.
hesion ligand for integrin a3b1 in epithelial basement 37. Cohen S. The stimulation of epidermal proliferation by a
membranes. Cell 1991: 65: 599–610. specific protein (EGF). Dev Biol 1965: 12: 394–407.

144
Cell biology of gingival wound healing

38. Colby RC. The general practitioner’s perspective of the cell contraction forces for collagen lattice contraction: an
etiology, prevention, and treatment of dry socket. Gen in vitro model of wound contraction. Tissue Cell 1990: 22:
Dent 1997: 45: 461–467, 471–472. 407–417.
39. Crawford SE, Stellmach V, Murphy-Ullrich JE, Ribeiro SM, 55. Epstein JB, Scully C. The role of saliva in oral health and
Lawler J, Hynes RO, Boivin GP, Bouck N. Thrombospond- the causes and effects of xerostomia. J Can Dent Assoc
in-1 is a major activator of TGF-b1 in vivo. Cell 1998: 93: 1992: 58: 217–21.
1159–1170. 56. Estes JM, Vande Berg JS, Adzick NS, MacGillivray TE, De-
40. Crossin KL. Tenascin: a multifunctional extracellular ma- smouliere A, Gabbiani G. Phenotypic and functional fea-
trix protein with a restricted distribution in development tures of myofibroblasts in sheep fetal wounds. Differen-
and disease. J Cell Biochem 1996: 61: 592–598. tiation 1994: 56: 173–181.
41. Cuvillier O, Pirianov G, Kleuser B, Vanek PG, Coso OA. 57. Fazakerley M, Field EA. Dry socket: a painful post-extrac-
Suppression of ceramide-mediated programmed cell tion complication. Dent Update 1991: 18: 31–34.
death by sphingosine-1-phosphate. Nature 1996: 381: 58. Ffrench-Constant C, Hynes RO. Patterns of fibronectin
800–803. gene expression and splicing during cell migration in
42. Darby I, Skalli O, Gabbiani G. a-smooth muscle actin is chicken embryos. Development 1988: 104: 369–382.
transiently expressed by myofibroblasts during experi- 59. Ffrench-Constant C, Hynes RO. Alternative splicing of
mental wound healing. Lab Invest 1990: 63: 21–29. fibronectin is temporally and spatially regulated in the
43. DeMali KA, Balciunaite E, Kazlauskas A. Integrins en- chicken embryo. Development 1989: 106: 375–388.
hance platelet-derived growth factor (PDGF)-dependent 60. Ffrench-Constant C, Van de Water L, Dvorak HF, Hynes
responses by altering the signal relay enzymes that are RO. Reappearance of an embryonic pattern of fibronectin
recruited to the PDGF b receptor. J Biol Chem 1999: 274: splicing during wound healing in the adult rat. J Cell Biol
19551–19558. 1989: 109: 903–914.
44. DeSimone DW, Norton PA, Hynes RO. Identification and 61. Field FK, Kerstein MD. Overview of wound healing in a
characterization of alternatively spliced fibronectin moist environment. Am J Surg 1994: 167: 2S–6S.
mRNAs expressed in early Xenopus embryos. Dev Biol 62. Firth JD, Putnins EE, Larjava H, Uitto VJ. Bacterial phos-
1992: 149: 357–369. pholipase C upregulates matrix metalloproteinase ex-
45. Desmouliere A, Badid C, Bochaton-Piallat ML, Gabbiani pression by cultured epithelial cells. Infect Immun 1997:
G. Apoptosis during wound healing, fibrocontractive dis- 65: 4931–4936.
eases and vascular wall injury. Int J Biochem Cell Biol 63. Fischer D, Brown-Ludi M, Schulthess T, Chiquet-Ehris-
1997: 1: 19–30. mann R. Concerted action of tenascin-C domains in cell
46. Desmouliere A, Gabbiani G. Modulation of fibroblastic adhesion, anti-adhesion and promotion of neurite out-
cytoskeletal features during pathological situations: the growth. J Cell Sci 1997: 10: 1513–1522.
role of extracellular matrix and cytokines. Cell Motil Cyto- 64. Fischer D, Tucker RP, Chiquet-Ehrismann R, Adams JC.
skel 1994: 29: 195–203. Cell-adhesive responses to tenascin-C splice variants in-
47. Desmouliere A, Geinoz A, Gabbiani F, Gabbiani G. Trans- volve formation of fascin microspikes. Mol Biol Cell 1997:
forming growth factor-b1 induces a-smooth muscle actin 8: 2055–2075.
expression in granulation tissue myofibroblasts and in 65. Fleischmajer R, Perlish JS, MacDonald ED 2nd, Schechter
quiescent and growing cultured fibroblasts. J Cell Biol A, Murdoch AD, Iozzo RV, Yamada Y. There is binding of col-
1993: 122: 103–111. lagen IV to b1 integrin during early skin basement mem-
48. Desmouliere A, Redard M, Darby I, Gabbiani G. Apoptosis brane assembly. Ann N Y Acad Sci 1998: 857: 212–227.
mediates the decrease in cellularity during the transition 66. Fluck J, Querfeld C, Cremer A, Niland S, Krieg T, Sollberg
between granulation tissue and scar. Am J Pathol 1995: S. Normal human primary fibroblasts undergo apoptosis
146: 56–66. in three-dimensional contractile collagen gels. J Invest
49. DiPersio CM, Hodival-Dilke KM, Jaenisch R, Kreidberg JA, Dermatol 1998: 110: 153–157.
Hynes RO. a3b1 Integrin is required for normal develop- 67. Friedl P, Zanker KS, Brocker EB. Cell migration strategies
ment of the epidermal basement membrane. J Cell Biol in 3-D extracellular matrix: differences in morphology,
1997: 137: 729–742. cell matrix interactions, and integrin function. Microsc
50. DiPietro LA, Nissen NN, Gamelli RL, Koch AE, Pyle JM, Res Tech 1998: 43: 369–378.
Polverini PJ. Thrombospondin 1 synthesis and function 68. Funato N, Moriyama K, Shimokawa H, Kuroda T. Basic
in wound repair. Am J Pathol 1996: 148: 1851–1860. fibroblast growth factor induces apoptosis in myofibro-
51. Dugina V, Alexandrova A, Chaponnier C, Vasiliev J, Gabbi- blastic cells isolated from rat palatal mucosa. Biochem
ani G. Rat fibroblasts cultured from various organs exhibit Biophys Res Commun 1997: 240: 21–26.
differences in a-smooth muscle actin expression, cyto- 69. Gabbiani G, Ryan GB, Majne G. Presence of modified
skeletal pattern, and adhesive structure organization. Exp fibroblasts in granulation tissue and their possible role in
Cell Res 1998: 238: 481–490. wound contraction. Experientia 1971: 27: 549–550.
52. Dvorak HF, Nagy JA, Feng D, Brown LF, Dvorak AM. Vascu- 70. Gailit J, Clark RA. Studies in vitro on the role of av and
lar permeability factor/vascular endothelial growth factor b1 integrins in the adhesion of human dermal fibroblasts
and the significance of microvascular hyperpermeability to provisional matrix proteins fibronectin, vitronectin,
in angiogenesis. Curr Top Microbiol Immunol 1999: 237: and fibrinogen. J Invest Dermatol 1996: 106: 102–108.
97–132. 71. Gailit J, Clarke C, Newman D, Tonnesen MG, Mosesson
53. Edgar WM. Saliva: its secretion, composition and func- MW, Clark RA. Human fibroblasts bind directly to fi-
tions. Br Dent J 1992: 172: 305–312. brinogen at RGD sites through integrin avb3. Exp Cell Res
54. Ehrlich HP, Rajaratnam JB. Cell locomotion forces versus 1997: 232: 118–126.

145
Häkkinen et al.

72. Gailit J, Pierschbacher M, Clark RA. Expression of func- 90. Gullberg D, Tingstrom A, Thuresson AC, Olsson L, Terra-
tional a4b1 integrin by human dermal fibroblasts. J Invest cio L, Borg TK, Rubin K. b1 integrin-mediated collagen
Dermatol 1993: 100: 323–328. gel contraction is stimulated by PDGF. Exp Cell Res 1990:
73. Gailit J, Welch MP, Clark RA. TGF-b1 stimulates expression 186: 264–272.
of keratinocyte integrins during re-epithelialization of cu- 91. Haapasalmi K, Zhang K, Tonnesen M, Olerud J, Sheppard
taneous wounds. J Invest Dermatol 1994: 103: 221–227. D, Salo T, Kramer R, Clark RAF, Uitto V-J, Larjava H. Kera-
74. Gailit J, Xu J, Bueller H, Clark RA. Platelet-derived growth tinocytes in human wounds express avb6 integrin. J In-
factor and inflammatory cytokines have differential ef- vest Dermatol 1996: 106: 42–48.
fects on the expression of integrins a1b1 and a5b1 by hu- 92. Häkkinen L, Heino J, Koivisto L, Larjava H. Altered inter-
man dermal fibroblasts in vitro. J Cell Physiol 1996: 169: action of human granulation-tissue fibroblasts with
281–289. fibronectin is regulated by a5b1 integrin. Biochim Bio-
75. Gardner H, Broberg A, Pozzi A, Laato M, Heino. Absence phys Acta 1994: 1224: 33–42.
of integrin a1b1 in the mouse causes loss of feedback 93. Häkkinen L, Larjava H. Characterization of fibroblast
regulation of collagen synthesis in normal and wounded clones from periodontal granulation tissue in vitro. J Dent
dermis. J Cell Sci 1999: 112: 263–272. Res 1992: 71: 1901–1907.
76. Giancotti FG. Integrin signalling: specificity and control 94. Häkkinen L, Westermarck J, Kahari VM, Larjava H. Hu-
of cell survival and cell cycle progression. Curr Opin Cell man granulation-tissue fibroblasts show enhanced
Biol 1997: 9: 691–700. proteoglycan gene expression and altered response to
77. Gibbons RJ, Etherden I. Fibronectin-degrading enzymes TGF-b1. J Dent Res 1996: 75: 1767–1778.
in saliva and their relation to oral cleanliness. J Peri- 95. Hashimoto-Uoshima M, Yan YZ, Schneider G, Aukhil I.
odontal Res 1986: 21: 386–395. The alternatively spliced domains EIIIB and EIIIA of hu-
78. Goldfinger LE, Hopkinson SB, deHart GW, Collawn S, Co- man fibronectin affect cell adhesion and spreading. J Cell
uchman JR, Jones JC. The a3 laminin subunit, a6b4 and Sci 1997: 110: 2271–2280.
a3b1 integrin coordinately regulate wound healing in cul- 96. Hemler ME. Integrin associated proteins. Curr Opin Cell
tured epithelial cells and in the skin. J Cell Sci 1999: 112: Biol 1998: 10: 578–585.
2615–2629. 97. Hertle MD, Kubler M-D, Leigh IM, Watt FM. Aberrant
79. Gould TR, Melcher AH, Brunette DM. Migration and divi- integrin expression during epidermal wound healing and
sion of progenitor cell populations in periodontal liga- in psoriatic epidermis. J Clin Invest 1992: 89: 1892–1901.
ment after wounding. J Periodontal Res 1980: 15: 20–42. 98. Hill SJ, Ebersole JL. The effect of lipopolysaccharide on
80. Goulhen F, Hafezi A, Uitto VJ, Hinode D, Nakamura R, growth factor-induced mitogenesis in human gingival
Grenier D, Mayrand D. Subcellular localization and cyto- fibroblasts. J Periodontol 1996: 67: 1274–1280.
toxic activity of the GroEL-like protein isolated from 99. Hormia M, Thesleff I, Perheentupa J, Pesonen K, Saxen L.
Actinobacillus actinomycetemcomitans. Infect Immun Increased rate of salivary epidermal growth factor secre-
1998: 66: 5307–5313. tion in patients with juvenile periodontitis. Scand J Dent
81. Grande JP. Role of transforming growth factor-b in tissue Res 1993: 101: 138–144.
injury and repair. Proc Soc Exp Biol Med 1997: 214: 27– 100. Howe A, Aplin AE, Alahari SK, Juliano RL. Integrin signal-
40. ling and cell growth control. Curr Opin Cell Biol 1998: 10:
82. Greenhalgh DG. The role of growth factors in wound heal- 220–231.
ing. J Trauma 1996: 41: 159–167. 101. Howell TH, Martuscelli G, Oringer J. Polypeptide growth
83. Greenwood JA, Murphy-Ullrich JE. Signalling of de-ad- factors for periodontal regeneration. Curr Opin Peri-
hesion in cellular regulation and motility. Microsc Res odontol 1996: 3: 149–156.
Tech 1998: 43: 420–432. 102. Huhtala P, Humphries MJ, McCarthy JB, Tremble PM,
84. Greiling D, Clark RA. Fibronectin provides a conduit for Werb Z, Damsky CH. Cooperative signalling by a5b1 and
fibroblast transmigration from collagenous stroma into a4b1 integrins regulates metalloproteinase gene express-
fibrin clot provisional matrix. J Cell Sci 1997: 110: 861–870. ion in fibroblasts adhering to fibronectin. J Cell Biol 1995:
85. Grenier D, Chao G, McBride BC. Characterization of so- 129: 867–879.
dium dodecyl sulfate–stable Bacteroides gingivalis pro- 103. Humphreys-Beher MG, Macauley SP, Chegini N, van Sett-
teases by polyacrylamide gel electrophoresis. Infect Im- en G, Purushotham K, Stewart C, Wheeler TT, Schultz GS.
mun 1989: 57: 95–99. Characterization of the synthesis and secretion of trans-
86. Grinnell F. Fibroblasts, myofibroblasts, and wound con- forming growth factor-a from salivary glands and saliva.
traction. J Cell Biol 1994: 124: 401–404. Endocrinology 1994: 134: 963–970.
87. Grinnell F, Ho CH, Lin YC, Skuta G. Differences in the 104. Ihara S, Motobayashi Y, Nagao E, Kistler A. Ontogenetic
regulation of fibroblast contraction of floating versus transition of wound healing pattern in rat skin occurring
stressed collagen matrices. J Biol Chem 1999: 274: 918– at the fetal stage. Development 1990: 110: 671–680.
923. 105. Irwin CR, Myrillas T, Smyth M, Doogan J, Rice C, Schor
88. Grinnell F, Zhu M, Carlson MA, Abrams JM. Release of SL. Regulation of fibroblast-induced collagen gel contrac-
mechanical tension triggers apoptosis of human fibro- tion by interleukin-1b. J Oral Pathol Med 1998: 27: 255–
blasts in a model of regressing granulation tissue. Exp Cell 259.
Res 1999: 248: 608–619. 106. Ivarsson M, Sundberg C, Farrokhnia N, Pertoft H, Rubin
89. Gross J, Farinelli W, Sadow P, Anderson R, Bruns R. On the K, Gerdin B. Recruitment of type I collagen producing
mechanism of skin wound ‘‘contraction’’: a granulation cells from the microvasculature in vitro. Exp Cell Res
tissue ‘‘knockout’’ with a normal phenotype. Proc Natl 1996: 229: 336–349.
Acad Sci U S A 1995: 92: 5982–5986. 107. Iyer VR, Eisen MB, Ross DT, Schuler G, Moore T, Lee JCF,

146
Cell biology of gingival wound healing

Trent JM, Staudt LM, Hudson J Jr, Boguski MS, Lashkari hyperplastic, and neoplastic tissues: biologic and patho-
D, Shalon D, Botstein D, Brown PO. The transcriptional logic implications. Hum Pathol 1991: 22: 636–643.
program in the response of human fibroblasts to serum. 123. Koyama H, Raines EW, Bornfeldt KE, Roberts JM, Ross R.
Science 1999: 283: 83–87. Fibrillar collagen inhibits arterial smooth muscle prolifer-
108. Juhasz I, Murphy GF, Yan HC, Herlyn M, Albelda SM. ation through regulation of Cdk2 inhibitors. Cell 1996: 87:
Regulation of extracellular matrix proteins and integrin 1069–1078.
cell substratum adhesion receptors on epithelium during 124. Krawczyk WS, Wilgram GF. Hemidesmosome and desmo-
cutaneous human wound healing in vivo. Am J Pathol some morphogenesis during epidermal wound healing. J
1993: 43: 1458–1469. Ultrastruct Res 1973: 45: 93–101.
109. Juliano R. Cooperation between soluble factors and inte- 125. Kurkinen M, Vaheri A, Roberts PJ, Stenman S. Sequential
grin-mediated cell anchorage in the control of cell growth appearance of fibronectin and collagen in experimental
and differentiation. Bioessays 1996: 18: 911–917. granulation tissue. Lab Invest 1980: 43: 47–51.
110. Kainulainen T, Häkkinen L, Hamidi S, Larjava K, Kallioin- 126. Kyriakides TR, Leach KJ, Hoffman AS, Ratner BD,
en M, Peltonen J, Salo T, Larjava H, Oikarinen A. Essential Bornstein P. Mice that lack the angiogenesis inhibitor,
role of laminin-5 during re-epithelialization of wounds. J thrombospondin 2, mount an altered foreign body reac-
Histochem Cytochem 1998: 46: 353–360. tion characterized by increased vascularity. Proc Natl
111. Kane CJM, Hebda PA, Mansbridge JN, Hamawalt PC. Di- Acad Sci U S A 1999: 96: 4449–4454.
rect evidence for spatial and temporal regulation of trans- 127. Laato M, Lehtonen OP, Niinikoski J. Granulation tissue for-
forming growth factor-b1 expression during cutaneous mation in experimental wounds inoculated with Staphylo-
wound healing. J Cell Physiol 1991: 148: 157–173. coccus aureus. Acta Chir Scand 1985: 151: 313–318.
112. Kanehisa J, Doi S, Yamanaka T, Takeuchi H. Salivary 128. Laato M, Niinikoski J, Lundberg C, Gerdin B. Inflamma-
fibronectin in man: an immunoblotting, radioimmuno- tory reaction and blood flow in experimental wounds in-
assay and immunohistochemical study. Arch Oral Biol oculated with Staphylococcus aureus. Eur Surg Res 1988:
1991: 36:265–272. 20: 33–38.
113. Kaplony A, Zimmermann DR, Fischer RW, Imhof BA, Od- 129. Lamberts BL, Pederson ED, Bial JJ, Tombasco PK.
ermatt BF, Winterhalter KH, Vaughan L. Tenascin Mr Fibronectin levels of unstimulated saliva from naval re-
220,000 isoform expression correlates with corneal cell cruits with and without chronic inflammatory peri-
migration. Development 1991: 112: 605–614. odontal disease. J Clin Periodontol 1989: 16: 342–346.
114. Karring T, Nyman S, Gottlow J, Laurell L. Development 130. Larjava H. Metabolic change in cultured gingival fibro-
of the biological concept of guided tissue regeneration – blasts exposed to bacterial extracts. Stimulation of hyalu-
animal and human studies. Periodontol 2000 1993: 1: 26– ronic acid synthesis. J Periodontal Res 1984: 19: 230–237.
35. 131. Larjava H, Haapasalmi K, Salo T, Wiebe C, Uitto V-J. Kera-
115. Kilcullen JK, Ly QP, Chang TH, Levenson SM, Steinberg tinocyte integrins in wound healing and chronic in-
JJ. Nonviable Staphylococcus aureus and its peptidoglycan flammation of the human periodontium. Oral Dis 1996:
stimulate macrophage recruitment, angiogenesis, fibro- 2: 77–86.
plasia, and collagen accumulation in wounded rats. 132. Larjava H, Salo T, Haapasalmi K, Kramer RH, Heino J. Ex-
Wound Repair Regen 1998: 6: 149–156. pression of integrins and basement membrane compo-
116. Kim JP, Zhang K, Chen JD, Kramer RH, Woodley DT. Vi- nents by wound keratinocytes. J Clin Invest 1993: 92:
tronectin-driven human keratinocyte locomotion is me- 1425–1435.
diated by the avb5 integrin receptor. J Biol Chem 1994: 133. Larjava H, Uitto VJ. Effects of extracts from Bacteroides
269: 26926–26932. gingivalis, Bacteroides intermedius, and Bacteroides asac-
117. Kirchberg K, Lange TS, Klein EC, Jungtaubl H, Heinen G, charolyticus on the growth of fibroblast lines obtained
Meyer-Ingold W, Scharffetter-Kochanek K. Induction of b1 from healthy and inflamed human gingiva. Oral Microbiol
integrin synthesis by recombinant platelet-derived Immunol 1987: 2: 112–116.
growth factor (PDGF-AB) correlates with an enhanced mi- 134. Larjava H, Uitto VJ, Eerola E, Haapasalo M. Inhibition of
gratory response of human dermal fibroblasts to various gingival fibroblast growth by Bacteroides gingivalis. Infect
extracellular matrix proteins. Exp Cell Res 1995: 220: 29– Immun 1987: 55: 201–205.
35. 135. Lee W, Aitken S, Sodek J, McCulloch CA. Evidence of a
118. Koivisto L, Larjava K, Häkkinen L, Uitto V-J, Heino J, Lar- direct relationship between neutrophil collagenase activ-
java H. Different integrins mediate cell spreading, hapto- ity and periodontal tissue destruction in vivo: role of ac-
taxis and lateral migration of HaCat keratinocytes on tive enzyme in human periodontitis. J Periodontal Res
fibronectin. Cell Adhes Commun 1999: 7: 245–257. 1995: 30: 23–33.
119. Konturek JW, Bielanski W, Konturek SJ, Bogdal J, Oleksy J. 136. Levenson SM, Kan-Gruber D, Gruber C, Molnar J, Seifter
Distribution and release of epidermal growth factor in E. Wound healing accelerated by Staphylococcus aureus.
man. Gut 1989: 30: 1194–1200. Arch Surg 1983: 118: 310–320.
120. Koseki N, Sato H, Yoshizato K. Suppression of growth-as- 137. Levine JH, Moses HL, gold LI, Nanney LMB. Spatial and
sociated phosphorylation of proteins of fibroblasts by col- temporal patters of immunoreactive transforming growth
lagen fibrils. Cell Adhes Commun 1996: 3: 463–474. factor b1, b2 and b3 during excitional wound repair. Am
121. Koseki N, Yoshizato K. Collagen-induced changes in the J Pathol 1993: 143: 368–380.
pattern of protein synthesis of fibroblasts. Cell Adhes 138. Lightner VA, Gumkowski F, Bigner DD, Erickson HP. Tena-
Commun 1994: 1: 355–366. scin/hexabrachion in human skin: biochemical identifi-
122. Koukoulis GK, Gould VE, Bhattacharyya A, Gould JE, cation and localization by light and electron microscopy.
Howeedy AA, Virtanen I. Tenascin in normal, reactive, J Cell Biol 1989: 108: 2483–2493.

147
Häkkinen et al.

139. Lin YC, Grinnell F. Decreased level of PDGF-stimulated Zentella A. Transforming growth factor-b. Cancer Surv
receptor autophosphorylation by fibroblasts in mechanic- 1992: 12: 81–103.
ally relaxed collagen matrices. J Cell Biol 1993: 122: 663– 156. Matsuoka Y, Spring J, Ballmer-Hofer K, Hofer U, Chiquet-
672. Ehrismann R. Differential expression of tenascin splicing
140. Lorimier S, Bouthors S, Droulle C, Maquin DL, Maquart variants in the chick gizzard and in cell cultures. Cell Dif-
FX, Gillery P, Emonard H Hornebeck W. The rate of fi- fer Dev 1990: 32: 417–423.
brinolysis is increased by free retraction of human gingi- 157. McClain SA, Simon M, Jones E, Nandi A, Gailit JO,
val fibroblast populated fibrin lattices. Int J Biochem Cell Tonnesen MG, Newman D, Clark RA Mesenchymal cell
Biol 1997: 29: 181–189. activation is the rate-limiting step of granulation tissue
141. Lorimier S, Gillery P, Hornebeck W, Chastang F, Laurent- induction. Am J Pathol 1996: 149: 1257–1270.
Maquin D, Bouthors S, Droulle C, Potron G, Maquart FX. 158. McCulloch CAG. Progenitor cell populations in the peri-
Tissue origin and extracellular matrix control neutral pro- odontal ligament of mice. Anat Rec 1985: 211: 258–262.
teinase activity in human fibroblast three-dimensional 159. Mio T, Adachi Y, Romberger DJ, Ertl RF, Rennard SI. Regu-
cultures. J Cell Physiol 1996: 168: 188–198. lation of fibroblast proliferation in three-dimensional col-
142. Lorimier S, Hornebeck W, Godeau G, Pellat B, Gillery P, lagen gel matrix in vitro. Cell Dev Biol Anim 1996: 32:
Maquart FX, Laurent-Maquin D. Morphometric studies of 427–433.
collagen and fibrin lattices contracted by human gingival 160. Miyamoto S, Katz BZ, Lafrenie RM, Yamada KM.
fibroblasts; comparison with dermal fibroblasts. J Dent Fibronectin and integrins in cell adhesion, signalling, and
Res 1998: 77: 1717–1729. morphogenesis. Ann N Y Acad Sci 1998: 857: 119–129.
143. Lukashev ME, Werb Z. ECM signalling: orchestrating cell 161. Miyamoto S, Teramoto H, Gutkind JS, Yamada KM. Inte-
behaviour and misbehaviour. Trends Cell Biol 1998: 8: grins can collaborate with growth factors for phos-
437–441. phorylation of receptor tyrosine kinases and MAP kinase
144. Mackie EJ, Halfter W, Liverani D. Induction of tenascin in activation: roles of integrin aggregation and occupancy of
healing wounds. J Cell Biol 1988: 107: 2757–2767. receptors. J Cell Biol 1996: 135: 1633–1642.
145. Magnuson VL, Young M, Schattenberg DG, Mancini MA, 162. Mochitate K, Pawelek P, Grinnell F. Stress relaxation of
Chen DL, Steffensen B, Klebe RJ. The alternative splicing contracted collagen gels: disruption of actin filament
of fibronectin pre-mRNA is altered during aging and in bundles, release of cell surface fibronectin and down-
response to growth factors. J Biol Chem 1991: 266: 14654– regulation of DNA and protein synthesis. Exp Cell Res
14662. 1991: 193: 198–207.
146. Mahaffey KW, Harrington RA, Simoons ML, Granger CB, 163. Moghal N, Sternberg PW. Multiple positive and negative
Graffagnino C, Alberts MJ, Laskowitz DT, Miller JM, Sloan regulators of signalling by the EGF-receptor. Curr Opin
MA, Berdan LG, MacAulay CM, Lincoff AM, Deckers J, To- Cell Biol 1999: 11: 190–196.
pol EJ, Califf RM. Stroke in patients with acute coronary 164. Montesano R, Orci L. Transforming growth factor b stimu-
syndromes: incidence and outcomes in the platelet glyco- lates collagen-matrixm contraction by fibroblasts: impli-
protein IIb/IIIa in unstable angina. Receptor suppression cations for wound healing. Proc Natl Acad Sci U S A 1988:
using integrilin therapy (PURSUIT) trial. The PURSUIT 85: 4894–4897.
Investigators. Circulation 1999: 99: 2371–2377. 165. Moore WE, Moore LV. The bacteria of periodontal dis-
147. Mäkelä M, Larjava H, Sorsa T, Uitto V-J. Matrix metallo- eases. Periodontol 2000 1994: 5: 66–77.
proteinase 2 (gelatinase A) is related to migration of kera- 166. Munger JS, Harpel JG, Giancotti FG, Rifkin DB. Interac-
tinocytes. Exp Cell Res 1999: 251: 67–78. tions between growth factors and integrins: latent forms
148. Mäkelä M, Salo T, Larjava H. MMP-9 from TNFa-stimu- of transforming growth factor-b are ligands for the inte-
lated keratinocytes binds to cell surface and type I colla- grin avb1. Mol Biol Cell 1998: 9: 2627–2638.
gen. Biochem Biophys Res Commun 1998: 253: 325–335. 167. Munger JS, Huang X, Kawakatsu H, Griffiths MJ, Dalton
149. Mäkelä M, Salo T, Uitto VJ, Larjava H. Matrix metallopro- SL, Wu J, Pittet JF, Kaminski N, Garat C, Matthay MA, Rif-
teinases (MMP-2 and MMP-9) of the oral cavity: cellular kin DB, Sheppard D. The integrin avb6 binds and acti-
origin and relationship to periodontal status. J Dent Res vates latent TGFb1: a mechanism for regulating pulmon-
1994: 73: 1397–1406. ary inflammation and fibrosis. Cell 1999: 96: 319–328.
150. Manabe R, Ohe N, Maeda T, Fukuda T, Sekiguchi K J. 168. Murphy-Ullrich JE, Lane TF, Pallero MA, Sage EH. SPARC
Modulation of cell-adhesive activity of fibronectin by the mediates focal adhesion disassembly in endothelial cells
alternatively spliced EDA segment. J Cell Biol 1997: 139: through a follistatin-like region and the Ca(2π)-binding
295–307. EF-hand. J Cell Biochem 1995: 57: 341–350.
151. Manabe R, Ohe N, Sekiguchi K. Alternatively spliced EDA 169. Murphy-Ullrich JE, Lightner VA, Aukhil I, Yan YZ, Erickson
segment regulates fibronectin-dependent cell cycle pro- HP, Hook M. Focal adhesion integrity is downregulated by
gression and mitogenic signal transduction. J Biol Chem the alternatively spliced domain of human tenascin. J Cell
1999: 274: 5919–5924. Biol 1991: 115: 1127–1136.
152. Mansbridge JN, Hanawalt PC. Role of transforming 170. Nagler RM, Kitrossky N, Chevion M. Antioxidant activity
growth factor-b in the maturation of human epidermal of rat parotid saliva. Arch Otolaryngol Head Neck Surg
keratinocytes. J Invest Dermatol 1988: 90: 336–341. 1997: 123: 989–993.
153. Martin P. Wound healing-aiming for perfect skin re- 171. Narani N, Arora PD, Lew A, Luo L, Glogauer M, Ganss B,
generation. Science 1997: 276: 75–81. McCulloch CA. Transforming growth factor-b induction of
154. Martin P, Lewis J. Actin cables and epidermal movement a-smooth muscle actin is dependent on the deformability
in embryonic wound healing. Nature 1992: 360: 179–183. of the collagen matrix. Curr Top Pathol 1999: 93: 47–60.
155. Massaque J, Cheifetz S, Laiho M, Ralph DA, Weis FM, 172. Nickoloff BJ, Mitra RS, Riser BL, Dixit VM, Varani J. Modu-

148
Cell biology of gingival wound healing

lation of keratinocyte motility. Correlation with produc- 189. Pöllänen MT, Salonen JI, Grenier D, Uitto V-J. Epithelial
tion of extracellular matrix molecules in response to cell response to challenge of bacterial lipoteichoic acids
growth promoting and antiproliferative factors. Am J and lipopolysaccharides in vitro. J Med Microbiol 2000:
Pathol 1988: 132: 543–551. 49: 1–8.
173. Niessen CM, Hogervorst F, Jaspars LH, de Melker AA, 190. Preissner KT. Structure and biological role of vitronectin.
Delwel GO, Hulsman EH, Kuikman I, Sonnenberg A. The Annu Rev Cell Biol 1991: 7: 275–310.
a6b4 integrin is a receptor for both laminin and kalinin. 191. Prieto AL, Edelman GM, Crossin KL. Multiple integrins
Exp Cell Res 1994: 211: 360–367. mediate cell attachment to cytotactin/tenascin. Proc Natl
174. Noguchi S, Ohba Y, Oka T. Effect of salivary epidermal Acad Sci U S A 1993: 90: 10154–10158.
growth factor on wound healing of tongue in mice. Am J 192. Prieto AL, Jones FS, Cunningham BA, Crossin KL, Edel-
Physiol 1991: 260: E620–E6205. man GM. Localization during development of alterna-
175. Obberghen-Schilling EV, Roche NS, Flanders KC, Sporn tively spliced forms of cytotactin mRNA by in situ hybridi-
MB, Roberts AB. Transforming growth factor b1 positively zation. J Cell Biol 1990: 111: 685–698.
regulates its own expression in normal and transformed 193. Prockop DJ. Marrow stromal cells as stem cells for non-
cells. J Cell Chem 1988: 263: 7741–7746. hematopoietic tissues. Science 1997: 276: 71–74.
176. Odland G, Ross R. Human wound repair. I. Epidermal re- 194. Rao RK, Thomas DW, Pepperl S, Porreca F. Salivary epider-
generation. J Cell Biol 1968: 39: 135–151. mal growth factor plays a role in protection of ileal mu-
177. Okada H, Murakami S. Cytokine expression in periodontal cosal integrity. Dig Dis Sci 1997: 42: 2175–2181.
health and disease. Crit Rev Oral Biol Med 1998: 9: 248– 195. Ravanti L, Heino J, Lopez-Otin C, Kahari VM. Induction
266. of collagenase-3 (MMP-13) expression in human skin
178. Oksala O, Salo T, Tammi R, Häkkinen L, Jalkanen M, Inki fibroblasts by three-dimensional collagen is mediated by
P, Larjava H. Expression of proteoglycans and hyaluronan p38 mitogen-activated protein kinase. J Biol Chem 1999:
during wound healing. J Histochem Cytochem 1995: 43: 274: 2446–2455.
125–135. 196. Richards DW, MacPhail LA, Dekker N, Greenspan D,
179. Oktay M, Wary KK, Dans M, Birge RB, Giancotti FG. Inte- Greenspan JS, Lozada-Nur F, Regezi JA. Expression of lam-
grin-mediated activation of focal adhesion kinase is re- inin 5, fibronectin, and epithelium-associated integrins in
quired for signalling to jun NH2-terminal kinase and pro- recurrent aphthous ulcers. J Dent Res 1996: 75: 1512–
gression through the G1 phase of the cell cycle. J Cell Biol 1517.
1999: 145: 1461–1470. 197. Riikonen T, Westermarck J, Koivisto L, Broberg A, Kahari
180. Oxford GE, Jonsson R, Olofsson J, Zelles T, Humphreys- VM, Heino J. Integrin a2b1 is a positive regulator of col-
Beher MG. Elevated levels of human salivary epidermal lagenase (MMP-1) and collagen a1(I) gene expression. J
growth factor after oral and juxtaoral surgery. J Oral Max- Biol Chem 1995: 270: 13548–13552.
illofac Surg 1999: 57: 154–158. 198. Robson MC, Stenberg BD, Heggers JP. Wound healing
181. Oxford GE, Nguyen KH, Alford CE, Tanaka Y, Humphreys- alterations caused by infection. Clin Plast Surg 1990: 17:
Beher MG. Elevated salivary EGF levels stimulated by 485–492.
periodontal surgery. J Periodontol 1998: 69: 479–484. 199. Romer J, Bugge TH, Pyke C, Lund LR, Flick MJ, Degen JL,
182. Pagani F, Zagato L, Vergani C, Casari G, Sidoli A, Baralle Dano. Impaired wound healing in mice with a disrupted
FE. Tissue-specific splicing pattern of fibronectin messen- plasminogen gene. Nat Med 1996: 2: 287–292.
ger RNA precursor during development and aging in rat. 200. Ronnov-Jessen L, Petersen OW. A function for filamen-
J Cell Biol 1991: 113: 1223–1229. tous-smooth muscle actin: retardation of motility in
183. Pereira RF, Halford KW, O’Hara MD, Leeper DB, Sokolov fibroblasts. J Cell Biol 1996: 134: 67–80.
BP, Pollard MD, Bagasra O, Prockop DJ. Cultured adherent 201. Ronnov-Jessen L, Petersen OW, Bissell MJ. Cellular
cells from marrow can serve as long-lasting precursor changes involved in conversion of normal to malignant
cells for bone, cartilage, and lung in irradiated mice. Proc breast: importance of the stromal reaction. Physiol Rev
Natl Acad Sci U S A 1995: 92: 4857–4861. 1996: 76: 69–125.
184. Phillips GR, Krushel LA, Crossin KL. Domains of tenascin 202. Rosenfeldt H, Lee DJ, Grinnell F. Increased c-fos mRNA
involved in glioma migration. J Cell Sci 1998: 111: 1095– expression by human fibroblasts contracting stressed col-
1104. lagen matrices. Mol Cell Biol 1998: 18: 2659–2667.
185. Phipps RP, Borrello MA, Blieden TM. Fibroblast heteroge- 203. Rousselle P, Lunstrum GP, Keene DR, Burgeson RE. Kal-
neity in the periodontium and other tissues. J Periodontal inin: an epithelium-specific basement membrane ad-
Res 1997: 32: 159–165. hesion molecule that is a component of anchoring fila-
186. Pilcher BK, Dumin JA, Sudbeck BD, Krane SM, Welgus ments. J Cell Biol 1991: 114: 567–576.
HG, Parks WC. The activity of collagenase-1 is required 204. Rubbia-Brandt L, Sappino AP, Gabbiani G. Locally applied
for keratinocyte migration on a type I collagen matrix. J GM-CSF induces the accumulation of a-smooth muscle
Cell Biol 1997: 137: 1445–1457. actin containing myofibroblasts. Virchows Arch 1991: 60:
187. Pilcher BK, Wang M, Qin XJ, Parks WC, Senior RM, Welgus 73–82.
HG. Role of matrix metalloproteinases and their inhi- 205. Ruoslahti E. Stretching is good for a cell. Science 1997:
bition in cutaneous wound healing and allergic contact 276: 1345–1346.
hypersensitivity. Ann N Y Acad Sci 1999: 878: 12–24. 206. Ruoslahti E. Fibronectin and its integrin receptors in can-
188. Pitaru S, McCulloch CA, Narayanan SA. Cellular origins cer. Adv Cancer Res 1999: 76: 1–20.
and differentiation control mechanisms during peri- 207. Saarialho-Kere UK, Kovacs SO, Pentland AP, Olerud JE,
odontal development and wound healing. J Periodontal Welgus HG, Parks WC. Cell-matrix interactions modulate
Res 1994: 29: 81–94. interstitial collagenase expression by human keratino-

149
Häkkinen et al.

cytes actively involved in wound healing. J Clin Invest 225. Sempowsky GD, Borello MA, Blieden TM, Barth RK,
1993: 92: 2858–2866. Phipps RP. Fibroblast heterogeneity in the healing wound.
208. Saga Y, Yagi T, Ikawa Y, Sakakura T, Aizawa S. Mice de- Wound Repair Regen 1995: 3: 120–131.
velop normally without tenascin. Genes Dev 1992: 6: 226. Serini G, Bochaton-Piallat ML, Ropraz P, Geinoz A, Borsi
1821–1831. L, Zardi L, Gabbiani G. The fibronectin domain ED-A is
209. Salo T, Lyons JG, Rahemtulla F, Birkedal-Hansen H, Larja- crucial for myofibroblastic phenotype induction by trans-
va H. Transforming growth factor-b1 up-regulates type IV forming growth factor-b1. J Cell Biol 1998: 142: 873–881.
collagenase expression in cultured human keratinocytes. 227. Shapiro SD. Matrix metalloproteinase degradation of
J Biol Chem 1991: 266: 11436–11441. extracellular matrix: biological consequences. Curr Opin
210. Salo T, Mäkelä M, Kylmäniemi M, Autio-Harmainen H, Cell Biol 1998: 10: 602–608.
Larjava H. Expression of MMP-2 and MMP-9 (72 kDa and 228. Shyy JY, Chien S. Role of integrins in cellular responses to
92 kDa type IV collagenases) during early human wound mechanical stress and adhesion. Curr Opin Cell Biol 1997:
healing. Lab Invest 1994: 70: 176–182. 9: 707–713.
211. Salonen J, Uitto VJ, Pan YM, Oda D. Proliferating oral epi- 229. Singer II, Scott S, Kawka DW, Kazazis DM, Gailit J, Ruos-
thelial cells in culture are capable of both extracellular lahti E. Cell surface distribution of fibronectin and vi-
and intracellular degradation of interstitial collagen. Ma- tronectin receptors depends on substrate composition ad
trix 1991: 11: 43–55. extracellular matrix accumulation. J Cell Biol 1988: 106:
212. Sappino AP, Schurch W, Gabbiani G. Differentiation reper- 2171–2182.
toire of fibroblastic cells: expression of cytoskeletal pro- 230. Skaleric U, Kramar B, Petelin M, Pavlica Z, Wahl SM.
teins as marker of phenotypic modulations. Lab Invest Changes in TGF-1 levels in gingiva, crevicular fluid and
1990: 63: 144–161. serum associated with periodontal inflammation in
213. Sarosiek J, Bilski J, Murty VL, Slomiany A, Slomiany BL. humans and dogs. Eur J Oral Sci 1997: 105: 136–142.
Role of salivary epidermal growth factor in the mainten- 231. Slavin J. The role of cytokines in wound healing. J Pathol
ance of physicochemical characteristics of oral and gas- 1996: 178: 5–10.
tric mucosal mucus coat. Biochem Biophys Res Commun 232. Sorsa T, Uitto VJ, Suomalainen K, Vauhkonen M, Lindy S.
1988: 152: 1421–1427. Comparison of interstitial collagenases from human gin-
214. Schiro JA, Chan BM, Roswit WT, Kassner PD, Pentland AP, giva, sulcular fluid and polymorphonuclear leukocytes. J
Hemler ME, Eisen AZ, Kupper TS. Integrin a2b1 (VLA-2) Periodontal Res 1988: 23: 386–393.
mediates reorganization and contraction of collagen mat- 233. Sottile J, Hocking DC, Swiatek PJ. Fibronectin matrix as-
rices by human cells. Cell 1991: 67: 403–410. sembly enhances adhesion-dependent cell growth. J Cell
215. Schmid P, Cox D, Bilbe G, McMaster G, Morrison C, Stähelin Sci 1998: 111: 2933–2943.
H, Lüscher N, Seiler W. TGF-bs and TGF-b type II receptor 234. Sporn MB, Roberts AB. Transforming growth factor-b: re-
in human epidermis: differential expression in acute and cent progress and new challenges. J Cell Biol 1993: 119:
chronic skin wounds. J Pathol 1993: 171: 191–197. 1017–1021.
216. Schmid P, Kunz S, Cerletti N, McMaster G, Cox D. Injury 235. Steffensen B, Duong AH, Milam SB, Potempa CL, Win-
induced expression of TGF-b1 is enhanced by exoge- born WB, Magnuson VL, Chen D, Zardeneta G, Klebe RJ.
nously applied TGF-bs. Biochem Biophys Res Commun Immunohistological localization of cell adhesion proteins
1993: 194: 399–406. and integrins in the periodontium. J Periodontol 1992: 63:
217. Schmitt-Graff A, Desmouliere A, Gabbiani G. Heteroge- 584–592.
neity of myofibroblast phenotypic features: an example of 236. Stenn KS, Malhotra R. Epithelialization. In: Cohe IK, Dieg-
fibroblastic cell plasticity. Virchows Arch 1994: 425: 3–24. elmann RF, Lindblad WJ, ed. Wound healing. Biochem-
218. Schneller M, Vuori K, Ruoslahti E. avb3 integrin associ- ical & clinical aspects. Philadelphia: WB Saunders, 1992:
ates with activated insulin and PDGF receptors and po- 115–127.
tentiates the biological activity of PDGF. EMBO J 1997: 16: 237. Stephens P, Davies KJ, al-Khateeb T, Shepherd JP, Thomas
5600–5607. DW. A comparison of the ability of intra-oral and extra-
219. Schor SL, Ellis I, Irwin CR, Banyard J, Seneviratne K, Dol- oral fibroblasts to stimulate extracellular matrix reorgan-
man C, Gilbert AD, Chisholm DM. Subpopulations of fe- ization in a model of wound contraction. J Dent Res 1996:
tal-like gingival fibroblasts: characterisation and potential 75: 1358–1364.
significance for wound healing and the progression of 238. Stephens P, Genever PG, Wood EJ, Raxworthy MJ. Integrin
periodontal disease. Oral Dis 1996: 2: 155–166. receptor involvement in actin cable formation in an in
220. Schultz G, Rotatori DS, Clark W. EGF and TGF-b in wound vitro model of events associated with wound contraction.
healing and repair. J Cell Biochem 1991: 45: 346–352. Int J Biochem Cell Biol 1997: 29: 121–128.
221. Schwartz MA, Baron V. Interactions between mitogenic 239. Stepp MA, Zhu L J. Upregulation of a9 integrin and tenas-
stimuli, or, a thousand and one connections. Curr Opin cin during epithelial regeneration after debridement in
Cell Biol 1999: 11: 197–202. the cornea. J Histochem Cytochem 1997: 45: 189–201.
222. Schwarzbauer JE. Alternative splicing of fibronectin: three 240. Stomski FC, Gani JS, Bates RC, Burns GF. Adhesion to
variants, three functions. Bioessays 1991: 13: 527–533. thrombospondin by human embryonic fibroblasts is me-
223. Sciubba JJ, Waterhouse JP, Meyer J. A fine structural com- diated by multiple receptors and includes a role for glyco-
parison of the healing of incisional wounds of mucosa protein 88 (CD36). Exp Cell Res 1992: 198: 85–92.
and skin. J Oral Pathol 1978: 7: 214–227. 241. Sundberg C, Ivarsson M, Gerdin B, Rubin K. Pericytes as
224. Sechler JL, Corbett SA, Wenk MB, Schwarzbauer JE. collagen-producing cells in excessive dermal scarring.
Modulation of cell-extracellular matrix interactions. Ann Lab Invest 1996: 74: 452–466.
N Y Acad Sci 1998: 857: 143–154. 242. Sundberg C, Rubin K. Stimulation of b1 integrins on

150
Cell biology of gingival wound healing

fibroblasts induces PDGF independent tyrosine phos- 259. Uitto VJ, Grenier D, Chan EC, McBride BC. Isolation of a
phorylation of PDGFb-receptors. J Cell Biol 1996: 132: chymotrypsinlike enzyme from Treponema denticola. In-
741–752. fect Immun 1988: 56: 2717–2722.
243. Taichman NS, Cruchley AT, Fletcher LM, Hagi-Pavli EP, 260. Uitto VJ, Haapasalo M, Laakso T, Salo T. Degradation of
Paleolog EM, Abrams WR, Booth V, Edwards RM, Mala- basement membrane collagen by proteases from some
mud D. Vascular endothelial growth factor in normal hu- anaerobic oral micro-organisms. Oral Microbiol Immunol
man salivary glands and saliva: a possible role in the 1988: 3: 97–102.
maintenance of mucosal homeostasis. Lab Invest 1998: 261. Uitto VJ, Larjava H. Extracellular matrix molecules and
78: 869–875. their receptors: an overview with special emphasis on
244. Taipale J, Saharinen J, Keski-Oja J. Extracellular matrix- periodontal tissues. Crit Rev Oral Biol Med 1991: 2: 323–
associated transforming growth factor-b: role in cancer 354.
cell growth and invasion. Adv Cancer Res 1998: 75: 87– 262. Uitto VJ, Larjava H, Heino J, Sorsa T. A protease of Bacter-
134. oides gingivalis degrades cell surface and matrix glyco-
245. Talonpoika J, Heino J, Larjava H, Häkkinen L, Paunio K. proteins of cultured gingival fibroblasts and induces se-
Gingival crevicular fluid fibronectin degradation in peri- cretion of collagenase and plasminogen activator. Infect
odontal health and disease. Scand J Dent Res 1989: 97: Immun 1989: 57: 213–218.
415–421. 263. Uitto VJ, Nieminen A, Coil J, Hurttia H, Larjava H. Oral
246. Tenovuo J. Antimicrobial function of human saliva – how fluid elastase as an indicator of periodontal health. J Clin
important is it for oral health? Acta Odontol Scand 1998: Periodontol 1996: 23: 30–37.
56: 250–256. 264. Uitto VJ, Raeste AM. Activation of latent collagenase of
247. Thesleff I, Partanen AM, Vainio S. Epithelial-mesenchy- human leukocytes and gingival fluid by bacterial plaque.
mal interactions in tooth morphogenesis: the roles of J Dent Res 1978: 57: 844–851.
extracellular matrix, growth factors, and cell surface re- 265. Uitto VJ, Suomalainen K, Sorsa T. Salivary collagenase.
ceptors. J Craniofac Genet Dev Biol 1991: 11: 229–237. Origin, characteristics and relationship to periodontal
248. Thesleff I, Viinikka L, Saxen L, Lehtonen E, Perheentupa health. J Periodontal Res 1990: 25: 135–142.
J. The parotid gland is the main source of human salivary 266. Vaalamo M, Weckroth M, Puolakkainen P, Kere J, Saarinen
epidermal growth factor. Life Sci 1988: 43: 13–18. P, Lauharanta J, Saarialho-Kere UK. Patterns of matrix
249. Thomson P. The microbiology of wounds. J Wound Care metalloproteinase and TIMP-1 expression in chronic and-
1998: 7: 477–478. normally healing human cutaneous wounds. Br J Derma-
250. Tingstrom A, Heldin CH, Rubin K. Regulation of fibro- tol 1996: 135: 52–59.
blast-mediated collagen gel contraction by platelet-de- 267. Van Waes C. Cell adhesion and regulatory molecules in-
rived growth factor, interleukin-1 and transforming volved in tumor formation, hemostasis, and wound heal-
growth factor-b1. J Cell Sci 1992: 102: 315–322. ing. Head Neck 1995: 17: 140–147.
251. Tomasek JJ, Akiyama SK. Fibroblast-mediated collagen gel 268. Varshney AC, Sharma DN, Singh M, Sharma SK, Nigam
contraction does not require fibronectin-a5b1 integrin in- JM. Therapeutic value of bovine saliva in wound healing:
teraction. Anat Rec 1992: 234: 153–160. a histomorphological study. Indian J Exp Biol 1997: 35:
252. Trachslin J, Koch M, Chiquet M. Rapid and reversible 535–537.
regulation of collagen XII expression by changes in tensile 269. Wary KK, Mainiero F, Isakoff SJ, Marcantonio EE, Giancot-
stress. Exp Cell Res 1999: 247: 320–328. ti FG. The adaptor protein Shc couples a class of integrins
253. Tremble P, Chiquet-Ehrismann R, Werb Z. The extracellu- to the control of cell cycle progression. Cell 1996: 87: 733–
lar matrix ligands fibronectin and tenascin collaborate in 743.
regulating collagenase gene expression in fibroblasts. Mol 270. Weinacker A, Chen A, Agrez M, Cone RI, Nishimura S,
Biol Cell 1994: 5: 439–453. Wayner E, Pytela R, Sheppard D. Role of integrin avb6 in
254. Tremble P, Damsky CH, Werb Z. Components of the nu- cell attachment to fibronectin: heterologous expression of
clear signalling cascade that regulate collagenase gene ex- intact and secreted forms of the receptor. J Biol Chem
pression in response to integrin-derived signals. Cell Biol 1994: 269: 6940–6948.
1995: 129: 1707–1720. 271. Welch MP, Odland GF, Clark RA. Temporal relationships
255. Tsao PW, Mousa SA. Thrombospondin mediates calcium of F-actin bundle formation, collagen and fibronectin
mobilization in fibroblasts via its Arg-Gly-Asp and car- matrix assembly, and fibronectin receptor expression to
boxyl-terminal domains. J Biol Chem 1995: 270: 23747– wound contraction. J Cell Biol 1990: 110: 133–145.
23753. 272. Werb Z, Tremble PM, Behrendtsen O, Crowley E, Damsky
256. Tuan TL, Song A, Chang S, Younai S, Nimni ME. In vitro CH. Signal transduction through the fibronectin receptor
fibroplasia: matrix contraction, cell growth, and collagen induces collagenase and stromelysin gene expression. J
production of fibroblasts cultured in fibrin gels. Exp Cell Cell Biol 1989: 109: 877–889.
Res 1996: 223: 127–134. 273. Westermarck J, Kahari VM. Regulation of matrix metallo-
257. Tynelius-Bratthall G. Crevicular and salivary fibronectin proteinase expression in tumor invasion. FASEB J 1999:
before and after gingivitis treatment. J Clin Periodontol 13: 781–792.
1988: 15: 283–287. 274. Whitby DJ, Ferguson MW. The extracellular matrix of lip
258. Uitto VJ, Airola K, Vaalamo M, Johansson N, Putnins wounds in fetal, neonatal and adult mice. Development
EE, Firth JD, Salonen J, Lopez-Otin C, Saarialho-Kere U, 1991: 112: 651–668.
Kahari VM. Collagenase-3 (matrix metalloproteinase-13) 275. Whitby DJ, Longaker MT, Harrison MR, Adzick NS, Fergu-
expression is induced in oral mucosal epithelium dur- son MWJ. Rapid epithelialization of fetal wounds is as-
ing chronic inflammation. Am J Pathol 1998: 152: 1489– sociated with the early deposition of tenascin. J Cell Sci
1499. 1991: 99: 583–586.

151
Häkkinen et al.

276. Wikesjo UM, Selvig KA. Periodontal wound healing and 288. Yang L, Qiu CX, Ludlow A, Ferguson MW, Brunner G. Ac-
regeneration. Periodontol 2000 1999: 19: 21–39. tive transforming growth factor-beta in wound repair: de-
277. Woodley DT. Reepithelialization. In: Clark RAF, ed. The termination using a new assay. Am J Pathol 1999: 154:
molecular and cellular biology of wound repair. New York: 105–111.
Plenum Press, 1996: 339–354. 289. Yeo TK, Brown L, Dvorak HF. Alterations in proteoglycan
278. Woodley DT, Kalebec T, Banes AJ, Link W, Prunieras M, synthesis common to healing wounds and tumors. Am J
Liotta. Adult human keratinocytes migrating over nonvi- Pathol 1991: 138: 1437–1450.
able dermal collagen produce collagenolytic enzymes that 290. Yokosaki Y, Monis H, Chen J, Sheppard D. Differential ef-
degrade type I and type IV collagen. J Invest Dermatol fects of the integrins a9b1, avb3, avb6 on cell proliferative
1986: 86: 418–423. responses to tenascin. Roles of the b subunit extracellular
279. Wu C, Chung AE, McDonald JA. A novel role for a3b1 inte- and cytoplasmic domains. J Biol Chem 1996: 271: 24144–
grins in extracellular matrix assembly. J Cell Sci 1995: 108: 24150.
2511–2523. 291. Yokosaki Y, Palmer EL, Prieto AL, Crossin KL, Bourdon
280. Wu C, Hughes PE, Ginsberg MH, McDonald JA. Identifi- MA, Pytela R, Sheppard D. The integrin a9b1 mediates
cation of a new biological function for the integrin avb3: cell attachment to a non-RGD site in the third fibronectin
initiation of fibronectin matrix assembly. Cell Adhes Com- type III repeat of tenascin. J Biol Chem 1994: 269: 26691–
mun 1996: 4: 149–158. 26696.
281. Xu J, Clark RA. Extracellular matrix alters PDGF regulation 292. Zambruno G, Marchisio PC, Marconi A, Vaschieri C, Mel-
of fibroblast integrins. J Cell Biol 1996: 132: 239–249. chiori A, Giannetti A, De Luca M. Transforming growth
282. Xu J, Zutter MM, Santoro SA, Clark RA. PDGF induction factor-b1 modulates b1 and b5 integrin receptor and in-
of a2 integrin gene expression is mediated by protein ki- duces the de novo expression of the avb6 heterodimer in
nase C-zeta. J Cell Biol 1996: 134: 1301–1311. normal keratinocytes: Implications for wound healing. J
283. Xue W, Mizukami I, Todd RF III, Petty HR. Urokinase- Cell Biol 1995: 129: 853–865.
type plasminogen activator receptors associate with b1 293. Zelles T, Purushotham KR, Macauley SP, Oxford GE,
and b3 integrins of fibrosarcoma cells: dependence on Humphreys-Beher MG. Saliva and growth factors: the
extracellular matrix components. Cancer Res 1997: 57: fountain of youth resides in us all. J Dent Res 1995: 74:
1682–1689. 1826–1832.
284. Yamada KM, Clark RAF. Provisional matrix. In: Clark RAF, 294. Zhang, L, Goulhen F, Grenier D, Mayrand D, Uitto V-J.
ed. The molecular and cellular biology of wound repair. Effects of GroEL-like heat-stress protein of Actinobacillus
New York: Plenum Press, 1996: 51–93. actinomycetemcomitans on epithelial signal transduction.
285. Yamada Y, Kleinman HK. Functional domains of cell ad- J Dent Res 1999: 78: 137.
hesion molecules. Curr Opin Cell Biol 1992: 4: 819–823. 295. Zhong C, Chrzanowska-Wodnicka M, Brown J, Shaub A,
286. Yang J, Tyler LW, Donoff RB, Song B, Torio AJ, Gallagher Belkin AM, Burridge K. Rho-mediated contractility ex-
GT, Tsuji T, Elovic A, McBride J, Yung CM, Galli SJ, Weller poses a cryptic site in fibronectin and induces fibronectin
PF, Wong DT. Salivary EGF regulates eosinophil-derived matrix assembly. J Cell Biol 1998: 141: 539–551.
TGF-a expression in hamster oral wounds. Am J Physiol 296. Ravanti L, Häkkinen L, Larjava H, Saarialho-Kere U, Fos-
1996: 270: 191–202. chi M, Han J, Kähäri V-M. Transforming growth factor-
287. Yang JT, Hynes RO. Fibronectin receptor functions in em- b induces collagenase-3 (MMP-13) expression by human
bryonic cells deficient in a5b1 integrin can be replaced gingival fibroblast via p38 mitogen-activated protein ki-
by aV integrins. Mol Biol Cell 1996: 7: 1737–1748. nase. J Biol Chem 1999: 274: 37292–37300.

152

You might also like