Sie sind auf Seite 1von 7

Periodontology 2000, Vol.

22, 2000, 44–50 Copyright C Munksgaard 2000


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

Biology of wound healing


I KRAMUDDIN A UKHIL

Periodontal tissues represent a unique system in the based on two excellent reviews on wound healing
human body where epithelial, soft and mineralized that the readers are referred to for more details (10,
connective tissues come together to form a junction. 21). Since the biological principles of wound repair
This junction, referred to as the dentogingival junc- would almost be the same, no distinction will be
tion, is a complex structure, and maintenance of the made between the healing in different types of peri-
integrity of this junction is critical for the preser- odontal defects. It is hoped that this review will allow
vation of underlying bone and periodontal ligament. the extrapolation of some of the basic principles of
Unfortunately, with chronic inflammation associated wound healing to the various periodontal surgical
with periodontal diseases, the structure of this junc- wounds.
tion is lost. Hence, attempts to control the destruc- As per the classic description of wound healing,
tive effects of chronic periodontal diseases and to initially there is temporary repair characterized by
some extent regenerate the lost tissues would require the formation of a clot in the wounded tissues. In-
the re-establishment of a dentogingival junction. flammatory cells followed by fibroblasts and endo-
Conventional periodontal therapy, be it surgical or thelial cells then invade the clot to form a granu-
nonsurgical in nature, usually involves instrumen- lation tissue, while the epithelial cells migrate to
tation in the inflamed dentogingival complex. Such cover the denuded surfaces (or form a junction at
therapies result in wounding of the already inflamed the tooth interface). Finally, maturation of the heal-
periodontal tissues. Thus, the consequence of such ing tissue matrix is seen along with contraction or
therapeutic procedures depends largely on the cellu- scarring. It is important to mention that these vari-
lar and molecular events associated with wound ous phases of wound healing overlap somewhat in
healing. Many of the cellular and molecular events time. This oversimplified explanation of wound heal-
in healing of periodontal wounds are similar to those ing basically summarizes the events at large, but nu-
seen in wounds elsewhere in the body except that, in merous studies in the last two decades have shed
the periodontal wounds, there is a mineralized tissue light on some of the important cellular and molecu-
interface at the junction of epithelium and connec- lar mechanisms. It is hoped that this new knowledge
tive tissue. would some day allow therapeutic manipulation to
Immediately following periodontal surgical pro- enhance wound healing.
cedures, the tissues represent surgically wounded
sites and a cascade of cellular and molecular events
set in to initiate wound repair. Over the past two The fibrin clot and
decades, numerous advances have been made in inflammatory cells
understanding the biology of wound healing. At the
same time, advances in the biology of the peri- Injury to the blood vessels during surgery/peri-
odontal tissues have led to the development of surgi- odontal therapeutic procedure causes extravasation
cal procedures that facilitate regeneration of the of blood. A fibrin-rich clot formed by blood coagu-
periodontal tissues lost because of chronic inflam- lation and platelet aggregation plugs the cut blood
mation. The newly described principle of guided vessels and also serves to protect the denuded
tissue regeneration (2) for the surgical treatment of tissues temporarily (10). The clot itself consists of
periodontal lesions represents a classic example of platelets within a network of cross-linked fibrin
how basic knowledge of cell biology could be applied fibers along with plasma fibronectin, vitronectin and
to enhance the clinical outcomes of therapy. The thrombospondin (21). Among the important func-
purpose of this chapter is to examine some of the tions of the clot are its role as a reservoir of growth
current basic concepts of wound healing and is factors and cytokines that are released by the de-

44
Biology of wound healing

granulation of activated platelets and serving as a Table 1. The integrin family of adhesion
provisional matrix for cell migration. The growth fac- receptors
tors and cytokines present in the fibrin clot in fact
Integrins Ligands
might be providing the start signals for wound re-
a, b; a2b1 Fibrillar collagen, laminin
pair. At first, there is recruitment of inflammatory a3b1 Fibronectin, entactin, epilgrin,
cells to the wound site followed by epithelialization, laminin, denatured collagen
a4b1 Fibronectin, VCAM-1
granulation tissue formation and angiogenesis. a5b1 Fibronectin (RGD)
Neutrophils and monocytes are usually recruited a6b1; a7b1; a6b4 Laminin
a8b1 Fibronectin, vitronectin
initially by the signals present in the clot. The neu- a9b1 Tenascin
trophils cleanse the wound of foreign particles and avb1; avb5 Fibronectin, vitronectin
debris and bacteria. It is important to note that neu- avb3; a11bb3 Vitronectin (RGD); fibronectin,
trophils remove the bacterial debris through the re- fibrinogen, von Willenbrand factor,
thrombospondin, denatured
lease of enzymes and toxic oxygen products (10). collagen
Hence, increasing numbers of contaminating bac- avb6 Fibronectin, tenascin
teria in the wound increase the potential for neutro- a4b7 Fibronectin (IIIcs)
avb8 Vitronectin
phil-mediated tissue destruction. Neutrophils also aMb2 Factor X, fibrinogen
serve as a source of pro-inflammatory cytokines pro- aXb2 Fibrinogen
viding signals that activate adjacent fibroblasts and
keratinocytes (17). Neutrophil infiltration ceases
after a few days, and they are eventually phagocytos-
ed either by macrophages or fibroblasts (10). Peri- consider changes in the expression of integrins by
pheral blood monocytes that continue to be re- cells migrating into the wound.
cruited into the wound site become macrophages In the normal tissues, keratinocytes use the inte-
upon activation. Fibrin along with fibronectin in the grins a6b4 to bind to laminin in the basal lamina,
clot acts as a provisional matrix for the influx of and these integrins have intracellular links with the
monocytes and fibroblasts (5). Macrophages con- keratin cytoskeletal network (21). In preparation for
tinue the task of phagocytosing bacterial, cellular migration, the keratinocytes at the edge of the surgi-
and matrix debris in the wound. Growth factors and cal wound have to dissolve the hemidesmosome
cytokines are continually synthesized and secreted attachment and begin to express other integrins that
into the wound environment by macrophages. Thus, are more suitable for the wound environment (Fig.
the wound repair signals initiated by degranulating 1). The migrating keratinocytes will start expressing
platelets and neutrophils are maintained by macro- the integrins a5b1 and aVb6 (for binding to
phages. fibronectin and tenascin respectively), the integrins
aVb5 for binding to vitronectin and, finally, reorgan-
ize the distribution of the integrins a2b1 (collagen
Re-epithelialization of wounds receptor). This mobilization and expression of new
integrins facilitates adhesion of keratinocytes to ma-
In the normal gingival tissues, the basal layer of epi- trix molecules in the provisional matrix as well as the
thelium is attached to the basal lamina. The keratino- adjacent wound debris. This can explain the lag time
cytes use receptors on their surface, known as inte- between wounding and the initiation of epithelial
grins, to bind to laminin in the basal lamina. Integrins migration. It is generally believed that the basal cells
are a family of cell adhesion receptors that mediate provide the majority of migrating keratinocytes, al-
cell surface interactions with extracellular matrix and though there is some evidence that some of the sup-
in some cases with other cells (19, 24, 32). There are rabasal cells may also migrate (13). Once the mi-
approximately 20 members of this family of cell sur- gration of epithelial cells has begun, cells of the basal
face receptors, and each integrin is a heterodimer layer small distance away from the wound edge un-
made up of one a and one b subunit in a non-covalent dergo proliferation, providing an extra source of
complex (33). The current list of the combinations of basal cells. The apical-basal polarity of the basal cell
various integrins and their respective biological layer at the wound edge is lost, and instead pseudo-
ligands is presented in Table 1. Changes in the combi- podia are seen extending from their free basolateral
nations of the various a and b subunits offer the inte- sides into the wound. Not much is known about the
grins specificity for the ligand. Since migration of cells mechanisms that drive the epithelial cell migration,
is fundamental to wound healing, it is important to but candidates include chemotactic factors, active

45
Aukhil

Fig. 1. Diagrammatic illustration of some of the major plasma fibronectin, vitronectin and thrombospondin.
events during healing of dermal wounds and periodontal This fibrin clot also serves as a reservoir for many of the
surgical wounds. a. The dermal wound. b. A healing peri- growth factors listed in Table 2. This is followed by recruit-
odontal wound with infrabony defects. Zones A represent ment of inflammatory cells in zone B, resulting in the
the borders of the wound where the epithelium (basal phagocytosing of debris and bacteria. Again zone B serves
layer) will migrate into the wound during epithelializ- as a reservoir of growth factors and cytokines. Zones C
ation. At this edge, the cells will have to dissolve the hemi- represent the borders of the wounds formed by the con-
desmosome attachment, downregulate the expression of nective tissue. Here, fibroblasts and endothelial cells show
integrin receptors a6b4 (used to bind to laminin), and up- proliferation in response to specific growth signals they
regulate integrin receptors a5b1, aVb6 and aVb5 that are receive as wound healing progresses. Matrix degradation
suitable for adhesion to provisional matrix components. is seen in zones B and C in preparation for the migration
Growth factors epidermal growth factor, transforming of these cells into zone B. Finally, granulation tissue forms
growth factor-a, heparin-binding epidermal growth factor in zone B followed by contraction of the wound and ma-
and keratinocyte growth factor are involved in stimulating trix remodeling. In the case of periodontal wounds, the
the proliferation of the epithelial cells here. Zone B ini- apical part of zone B may be populated by cells originat-
tially represents the fibrin clot consisting of platelets ing from bone and periodontal ligament, while the more
within a network of cross-linked fibrin fibers along with coronal part of zone B may get epithelialized.

contact guidance, absence of neighboring cells or a epidermal growth factor, transforming growth factor-
combination of the above (10). Interestingly, epi- a, heparin-binding epidermal growth factor and
thelial cell migration does not appear to depend on keratinocyte growth factor. A list of growth factors
cell proliferation since molecules such as trans- active in the healing wounds, their possible sources
forming growth factor-b, despite being a potent in- and biological effects are presented in Table 2.
hibitor of keratinocyte proliferation, promote the
migration of epithelial cells in organ cultures (10).
Once re-epithelialization is complete, the compo- Matrix degradation and
nents of basal lamina are deposited in a sequential the wound-cleaning process
manner starting from the wound margin and the
epithelial cells revert to their normal phenotype. Migration of epithelial cells through the fibrin clot or
Several growth factors seem to be key players in along the junction between the clot and underlying
regulating the proliferation of keratinocytes in heal- connective tissue (mucoperiosteal flap surface in the
ing wounds. Among the key growth factors are the case of periodontal surgery) requires the creation of

46
Biology of wound healing

Table 2. Growth factors involved in wound healing


Growth factor Source Effect
Fibroblast growth factors 1, 2 and 4 Macrophages, endothelial cells Fibroblast proliferation and angiogenesis
Transforming growth factor-a Macrophages, keratinocytes Re-epithelialization
Transforming growth factors b1 and b2 Platelets, macrophages Fibroblast and macrophage chemotaxis;
extracellular matrix synthesis; secretion of
protease inhibitors
Epidermal growth factor Platelets Re-epithelialization
Platelet-derived growth factor Platelets, macrophages, Fibroblast and macrophage chemotaxis,
(isoforms AA, AB and BB) keratinocytes fibroblast proliferation, and matrix synthesis
Keratinocyte growth factor Dermal fibroblasts Keratinocyte proliferation
Insulin-like growth factor Plasma, platelets Endothelial and fibroblast proliferation
Vascular endothelial growth factor Keratinocytes, macrophages Angiogenesis
Interleukin 1a and b Neutrophils Activate growth factor expression in
macrophages, keratinocytes and fibroblasts
Tumor necrosis factor-a Neutrophils Activate growth factor expression in
macrophages, keratinocytes and fibroblasts

a migrating path. This is achieved by the dissolution mainly of new capillaries, macrophages, fibroblasts
of the fibrin barrier by the enzyme plasmin that is and some loose connective tissue. Needless to say,
derived from the activation of plasminogen in the granulation tissue is a complex reservoir of cytokines
clot. The two activators, tissue-type plasminogen ac- possessing chemoattractive, mitogenic and other
tivator and urokinase-type plasminogen activator regulatory activities (21). Growth factors are also
along with its receptor, are upregulated in the mi- present in the granulation tissue and, at this stage of
grating keratinocytes (21). The importance of plas- healing, are derived mostly from macrophages. De-
minogen in wound re-epithelialization is demon- pending on the stage of the granulation tissue and
strated by the lack of re-epithelialization of wounds the predominant cytokines, numerous activities are
in mice where the plasminogen gene has been seen occurring such as cell proliferation, chemotaxis
knocked out (21). In addition to the fibrinolytic en- and phenotypic expression of cells (27). It is, how-
zyme plasmin that causes lysis of fibrin, several ever, important to note that the activities of certain
other proteases are also expressed to clear the path cell types and the nature of the extracellular matrix
for cell migration. Matrix metalloproteinases (MMP) environment influence to a large extent the final
are a family of enzymes with each member of the phenotype expression by the cells.
family specifically cleaving a subset of matrix pro- During the formation of granulation tissue,
teins. MMP-1 (also known as interstitial collagenase) macrophages, fibroblasts and new blood vessels
is secreted by those basal cells that have gone past grow into the wound space in a coordinated manner
the free edge of the basal lamina. MMP-1 degrades (Fig. 1). Their interdependence is illustrated by the
native collagens and aids cell migration by destroy- release of cytokines by macrophages that stimulate
ing collagens I and III. Similarly, MMP-9 (also known fibroblasts to synthesize an extracellular matrix. This
as gelatinase B) can cleave the collagen in basal lam- extracellular matrix serves to support cell and vascu-
ina (type IV) and the collagen that forms the an- lar in-growth carrying nutrients to sustain the cellu-
choring fibrils (type VII), thereby releasing the basal lar functions (21). The term fibroplasia is used to re-
cells from their adhesion to the basal lamina. MMP- fer to that part of the granulation tissue made up
10 (also known as stromelysin-2) is also expressed in of predominantly fibroblasts and extracellular matrix
wounds and is thought to have a wide spectrum of they make. In addition to macrophages, fibroblasts
substrate specificity (21). themselves express many cytokines to which they
can also respond in an autocrine manner (27). Cyto-
kines induce proliferation of fibroblasts and regulate
Granulation tissue and the production of extracellular matrix. Similarly,
contraction of the wound growth factors are also involved in wound fibroplas-
ia. Wounds supplemented with purified growth fac-
Granulation tissue formation usually can begin tors show accelerated granulation tissue formation
around day 4 after the wounding and consists (20, 28).

47
Aukhil

The extracellular matrix and fibroblasts function fibronectin molecule may be facilitating the mi-
in a reciprocal manner during wound healing. Com- gration of cells by modulating the adhesiveness of
ponents of the extracellular matrix such as fibronec- the substrata (8, 15). Another spliced domain of
tin and collagen facilitate the adhesion and mi- fibronectin, the CS-III domain, also may be involved
gration of fibroblasts in the granulation tissue. Fibro- in the migration of cells in wounds. Fibroblasts use
blasts synthesize and decorate the extracellular the integrin receptor a4b1 (Table 1) to bind to the
matrix, and in turn the extracellular matrix regulates CS-III domain of fibronectin. Interestingly, it has
gene expression and the general behavior of fibro- been shown in cell culture studies that the express-
blasts (10). Fibroblasts adhere to fibronectin, colla- ion of the integrin a4b1 facilitates migration, while
gen and vitronectin via the integrin receptors listed the classic fibronectin receptor a5b1 that binds to
in Table 1. It is important to note that fibroblasts, the RGD sequence can retard movement (7, 14).
like the keratinocytes, have to rearrange their inte- Fibroblasts in healing wound have been known to
grin expression profiles in preparation for migration. over-express the spliced domain CS-III (5, 11). Once
In the normal tissues, fibroblasts reside in collagen- cell migration into wounds is completed, fibroblasts
rich matrices. In response to wounding, the fibro- show an increased expression of the classic a5b1
blasts in the vicinity of the wound have to downreg- integrin (29). Fibroblasts in wounds also use vi-
ulate the integrin receptors for collagen and up- tronectin and fibrin directly as substrata for ad-
regulate those needed for adhesion to components hesion during migration. This is facilitated by the
of the provisional matrix such as fibrin, fibronectin availability of cell membrane receptors for these ma-
and vitronectin. The ability of fibroblasts to respond trix proteins (Table 1). Finally, the migration of
to signals from the extracellular environment is re- fibroblasts in wounds is also stimulated indirectly by
markable. For example, when simultaneously chal- growth factors such as platelet-derived growth factor
lenged by signals from both the provisional matrix and transforming growth factor-b by upregulating
(fibrin or fibronectin) as well as growth factors (such some of the integrin receptors mentioned above (1,
as platelet-derived growth factor), fibroblasts re- 12).
spond by upregulating the receptors for provisional As wound healing progresses, the provisional ma-
matrix components. However, when challenged by trix is replaced by a new, collagen-rich matrix syn-
the same growth factor (platelet-derived growth fac- thesized by fibroblasts migrating into the wound.
tor) in the presence of a collagenous matrix, fibro- The synthesis of specific extracellular matrix mol-
blasts respond by upregulating the receptors for col- ecules by fibroblasts in the wound is regulated by
lagen and not the provisional matrix receptors (31). transforming growth factor-b1 and some other
Not much is known about the effects of extracellular growth factors listed in Table 2 (10, 21). Cytokines
matrix molecules in the wound environment on the such as interleukin-4 can also induce expression of
regulation of gene expression in fibroblasts. This is collagenous matrices in wounds (23). Once the re-
complicated by the fact that some of the extracellu- quired amount of collagenous matrix is synthesized,
lar matrix molecules occur in different isoforms due the signals that induce down-regulation of collagen
to alternative splicing of their transcripts at the pre– synthesis are not clearly known. It is possible that
messenger RNA level. For example, the well-known there may exist some kind of regulation by the colla-
molecule fibronectin occurs as different isoforms gen matrix itself (9). Around 7–10 days after
due to the splicing in or out of the alternatively wounding, some of the fibroblasts in the wound
spliced domains EIIIB and EIIIA in its gene structure. transform into myofibroblasts and express a-smooth
While the fibronectin from plasma does not contain muscle actin. Such transformation allows these myo-
either of the two spliced domains, the fibronectin fibroblasts to generate strong contractile forces that
synthesized by fibroblasts and macrophages in is responsible for wound contraction (21). In the fi-
wounds includes the spliced domains EIIIB and nal stages of fibroplasia, the number of fibroblasts
EIIIA. Remarkably, the EIIIB and EIIIA inclusive iso- and myofibroblasts in the healing wound are de-
form of fibronectin seen in healing wounds is the creased by programmed cell death. Compared with
predominant form of fibronectin in embryonic wounds in the adult tissues, embryonic wounds heal
tissues where cell migration is active (11). The ad- without much contraction and scarring. In the em-
vantages of including the spliced domains EIIIB and bryos, there is no conversion of fibroblasts to myo-
EIIIA in fibronectin during embryonic development fibroblasts and the angiogenic response is also of a
and wound healing are not clear. Recent studies have lesser magnitude. Compared with wounds in adult
suggested that inclusion of these domains in the tissues, wounds in embryos show low but transient

48
Biology of wound healing

levels of expression of transforming growth factor-b1 applied to manipulate the process of wound healing.
(30) and since transforming growth factor-b1 has For example, application of epidermal growth factor
been implicated in pathofibrotic conditions, this can and transforming growth factor-a to burn wounds
explain the tendency for adult wounds to show scar- in animal models has been shown to enhance re-
ring and contraction. Antibodies that neutralize the epithelialization (4, 25). Similarly, application of
effects of transforming growth factor-b1 in healing keratinocyte growth factor to skin wounds has been
wounds have been shown to reduce scarring when shown to have mitogenic effects on the healing epi-
delivered to wounds (26). thelium (22). Since cell adhesion and migration are
essential to wound healing, attempts have been
made in the past to use topical application of
Angiogenesis of wounds fibronectin in periodontal wound healing (18). How-
ever, using recombinant molecules as exogenous
Angiogenesis refers to the formation of new blood sources to supplement wound healing has to be ap-
vessels and is a crucial event during the healing of proached with caution. For example, in the case of
wounds. The term granulation tissue in wounds was fibronectin, the plasma form of fibronectin (without
coined in reference to the red granular appearance spliced domains EIIIB and EIIIA) is already available
of new blood vessels that invade the healing connec- in large quantities in the provisional matrix of a heal-
tive tissues. As listed in Table 2, several growth fac- ing wound. Supplementing the same has no biologi-
tors are important in the induction of angiogenesis cal rationale and enhanced effects should not be ex-
in healing wounds. Fibroblast growth factor-2 is syn- pected. The biological functions of the spliced do-
thesized by macrophages and damaged endothelial mains EIIIB, EIIIA and the CS-III segments are not
cells, while vascular endothelial growth factor is in- known. Should their roles be specific in enhancing
duced in wound-edge keratinocytes and macro- cell migration (15), specific isoforms of cell adhesion
phages. Experiments where fibroblast growth factor- molecules that allow weak cell adhesion but favor
2 is depleted show blockage of wound angiogenesis migration of cells would be useful, and studies to
(3). As in the case of fibroblasts and keratinocytes, this effect are currently in progress. The provisional
the endothelial cells also have to upregulate specific matrix is rich in growth factors in the normal,
integrins (aVb3) on their surface in order to respond healthy subjects. Systemic conditions such as dia-
to angiogenic signals. All the signals that induce pro- betes may be accompanied by reduction in the avail-
liferation, migration and phenotype expression in ability of some of the growth factors; in these cases,
endothelial cells have not been completely under- supplementing the appropriate growth factor may
stood yet. It is clear that angiogenesis is a complex be beneficial. Since the half-life for some of the
process and relies on the availability of an appropri- growth factors may be short, supplementing wounds
ate matrix in the wound. As endothelial cells migrate with the appropriate growth factors during the later
into the provisional matrix, they form tubes sur- stages of healing via some sort of delayed release
rounded by their own provisional matrix initially fol- mechanism could be useful in the healing of wounds
lowed by a true basement membrane. As in the case in healthy subjects as well. Preliminary data on the
of fibroblasts, endothelial cells involved with angio- use of recombinant growth factors during peri-
genesis in wounds also undergo programmed cell odontal surgery to treat osseous defects shows
death during the ultimate maturation of the matrix promising results (16).
characterized by regression of capillaries. Future research will have to be directed towards
understanding in more detail the molecular mech-
anisms of differential gene expression in healing
wounds. Wound healing is achieved by a series of
Applied clinical aspects and coordinated efforts by inflammatory cells, keratino-
future approaches to enhance cytes, fibroblasts and endothelial cells responding to
wound healing a complex array of signals. Many of these events
have start and stop signals. A thorough understand-
With the explosion in knowledge of growth factors, ing of these signals and their consequences (differ-
cell adhesion molecules and cytokines in the last two ential gene expression) in responder cells should
decades, understanding of the cellular and molecu- make possible, hopefully in the near future, thera-
lar biology of wound healing has improved signifi- peutic manipulation of wounds that leads to real re-
cantly. Some of this new knowledge is already being generation of the damaged tissues.

49
Aukhil

Acknowledgments 15. Hashimoto-Uoshima M, Yan YZ, Schneider G, Aukhil I. The


alternatively spliced domains EIIIB and EIIIA of human
fibronectin affect cell adhesion and spreading. J Cell Sci
This work was supported in part by a grant from the 1997: 110: 2271–2280.
National Institutes of Health (DE-07801). I thank 16. Howell TW, Fiorellini JP, Paquette DW, Offenbacher S, Gian-
Donna Yeager for assistance in the preparation of nobile WV, Lynch S. A phase I/II clinical trial to evaluate a
this manuscript. combination of recombinant human platelet derived
growth factor-BB and recombinant human insulin-like
growth factor-I in patients with periodontal disease. J Peri-
odontol 1997: 68: 1186–1193.
17. Hubner G, Branchle M, Smola H, Madlener M, Fassler R,
References Werner S. Differential regulation of pro-inflammatory cyto-
kines during wound healing in normal and glucocorticoid
1. Ahlen K, Rubin K. Platlet-derived growth factor-BB stimu- treated mice. Cytokine 1996: 8: 548–556.
lates synthesis of the integrin a2 subunit in human diploid 18. Hynes RO. Wound healing, inflammation and fibrosis. In:
fibroblasts. Exp Cell Res 1994: 215: 347–353. Fibronectins. Berlin: Springer-Verlag, 1990.
2. Aukhil I. Potential contributions of cellular and molecular 19. Hynes RO. Integrins: versatility, modulation, and signaling
biology to periodontal tissue regeneration. Curr Opin Dent in cell adhesion. Cell 1992: 69: 11–25.
1992: 2: 91–96. 20. Lynch SE, Colvin RB, Antoniades HN. Growth factors in
3. Broadley KN, Aquino AM, Woodward SC, Buckley-Sturrock wound healing. Single and synergistic effects on partial
A, Sato Y, Rifkin DB, Davidson JM. Monospecific antibodies thickness porcine skin wounds. J Clin Invest 1989: 84: 640–
implicate basic fibroblast growth factor in normal wound 646.
repair. Lab Invest 1989: 61: 571–575. 21. Martin P. Wound healing – aiming for perfect skin regenera-
4. Brown GL, Curtsinger L, Brightwell JR, Ackerman DM, Tob- tion. Science 1997: 276: 75–81.
in GR, Polk HC, George-Nascimento C, Valenzuela P, 22. Pierce GF, Yanagihara D, Klopchin K, Danilenko DM, Hsu
Schultz GS. Enhancement of epidermal regeneration by E, Kenney WC, Morris CF. Stimulation of all epithelial ele-
biosynthetic epidermal growth factor. J Exp Med 1986: 163: ments during skin regeneration by keratinocyte growth fac-
1319–1324. tor. J Exp Med 1994: 179: 831–840.
5. Brown LF, Dubin D, Lavigne L, Logan B, Dvorak HF, Van de 23. Postlethwaite AE, Holness MA, Katai H, Raghow R. Human
Water F. Macrophages and fibroblasts express embryonic fibroblasts synthesize elevated levels of extracellular matrix
fibronectins during cutaneous wound healing. Am J Pathol proteins in response to interleukin-4. J Clin Invest 1992: 90:
1993: 142: 793–801. 1479–1485.
6. Brown LF, Lanir N, McDonagh J, Tognazi K, Dvorak AM, 24. Ruoslahti E. Integrins. J Clin Invest 1991: 87: 1–5.
Dvorak HF. Fibroblast migration in fibrin gel matrices. Am 25. Schultz S, White M, Mitchell R, Brown G, Lynch J, Twardzik
J Pathol 1993: 142: 273–283. DR, Todaro GJ. Epithelial wound healing enhanced by
7. Chan BM, Kassner PD, Schiro JA, Byers R, Kupper TS, transforming growth factor alpha and vaccinia growth fac-
Hemler ME. Distinct cellular functions mediated by differ- tor. Science 1987: 235: 350–352.
ent VLA integrin a subunit cytoplasmic domains. Cell 1992: 26. Shah M, Foreman DM, Ferguson MWJ. Control of scarring
68: 1051–1060. in adult wounds by neutralizing antibodies to transforming
8. Chen W, Culp L. Adhesion mediated by fibronectin’s alter- growth factor-b. Lancet 1992: 339: 213–214.
natively spliced EIIIB and its neighboring type III repeats. 27. Sporn M, Roberts AM. Peptide growth factors and inflam-
Exp Cell Res 1996: 223: 9–19. mation, tissue repair, and cancer. J Clin Invest 1986: 78:
9. Clark RAF, Nielsen LD, Welch MP, McPherson JL. Collagen 329–332.
matrices attenuate the collagen synthetic response of cul- 28. Sporn M, Roberts AM, Shull JH, Smith JM, Ward JM, Sodek
tured fibroblasts to TGF-b. J Cell Sci 1995: 108: 1252–1261. J. Polypeptide transforming growth factor isolated from bo-
10. Clark RAF. Wound repair: overview and general consider- vine sources and used for wound healing in vitro. Science
ations. In: The molecular and cellular biology of wound re- 1983: 219: 1329–1331.
pair. 2nd edn. New York: Plenum Press, 1996. 29. Welch MP, Odland GF, Clark RAF. Temporal relationships of
11. French-Constant C, Van de Water L, Dvorak HF, Hynes RO. F-actin bundle formation, collagen and fibronectin matrix
Reappearence of an embryonic pattern of fibronectin spli- assembly, and fibronectin receptor exression to wound
cing during wound healing in the adult rat. J Cell Biol 1989: contraction. J Cell Biol 1990: 110: 133–145.
109: 903–914. 30. Whitby DJ, Ferguson MWJ. Immunohistochemical localiz-
12. Gailit J, Bueller H, Clark RAF. Platelet-derived growth factor ation of growth factors in fetal wound healing. Dev Biol
and inflammatory cytokines have differential effects on the 1991: 147: 207–215.
expression of integrins a1b1 and a5b1 by human dermal 31. Xu J, Clark RAF. Extracellular matrix alters platelet derived
fibroblasts in vitro. J Cell Physiol 1996: 169: 281–289. growth factor regulation of fibroblast integrins. J Cell Biol
13. Garlick JA, Taichman LB. Fate of human keratinocytes dur- 1996: 132: 239–249.
ing reepithelialization in an organotypic culture model. Lab 32. Yamada KM. Adhesive recognition sequences. J Biol Chem
Invest 1992: 70: 916–924. 1991: 266: 12809–12812.
14. Giancotti FG, Ruoslahti E. Elevated levels of the a5b1 33. Yamada KM, Gailit J, Clark RAF. Integrins in wound repair.
fibronectin receptor suppress the transformed phenotype In: Clark RAF, ed. The molecular and cellular biology of
of chinese hamster ovary cells. Cell 1990: 60: 849–859. wound repair. 2nd edn. New York: Plenum Press, 1996.

50

Das könnte Ihnen auch gefallen