<<

Periodontology 2000, Vol. 41, 2006, 196–217 Copyright Blackwell Munksgaard 2006 Printed in Singapore. All rights reserved 2000

Regeneration of periodontal tissues: revisited

MARGARITA ZEICHNER-DAVID

Virtually all types of are caused responsible for the attachment of teeth in the oral by periodontal pocket infections, although several cavityÕ (144). Several excellent reviews have been other factors, including trauma, aging, systemic dis- published describing the embryonic lineage of the eases, genetics, etc., can contribute to the destruction principal periodontal tissues (, periodontal of the (1, 18, 31, 52, 60, 107, 128, 127, , gingiva and alveolar ), as well as the 194). Repair of the periodontium and the regener- cells and components of the ation of periodontal tissues remains a major goal in periodontium (10, 13, 14, 21, 19, 46, 45, 51, 71, 80, 82, the treatment of periodontal disease and is an area 144, 158, 185, 186, 193, 212, 214, 243, 244, 245). still in need of major research attention, as recently Formation of the periodontium is initiated with stated by the American Academy of Periodontology the process of root formation where, following (260). In general, to achieve complete tissue regen- formation, the apical mesenchyme continues eration and repair, it is necessary to recapitulate the to proliferate to form the developing periodontium, process of embryogenesis and morphogenesis in- while the inner and outer enamel epithelia fuse volved in the original formation of the tissue. In the below the level of the cervical enamel to produce a case of the periodontium, complete periodontal re- bilayered epithelial sheath, termed the Hertwig’s pair entails de novo cementogenesis, osteogenesis . As these cells divide, there is and the formation of periodontal ligament fibers. an apical migration of the Hertwig’s epithelial root Current strategies for periodontal repair are based on sheath cells through the underlying dental ectome- anti-infectious measures such as scaling and root senchymal tissues, dividing them into the dental planing, guided tissue regeneration (with or without papilla and the (Fig. 1). As the root bone grafts) or the use of growth factors, none of develops, the first radicular mantle is formed which fully restore the architecture of the original and the epithelial sheath is fenestrated. It is believed periodontium. Several different approaches involving that cells of the Hertwig’s epithelial root sheath tissue engineering are currently being explored to migrate away from the root into the region of the achieve complete, reliable and reproducible regen- future periodontal ligament where they re-associate eration of the periodontium. As tissue engineering is to form the Epithelial Rest of Malassez. However, defined as the science that develops techniques not all Hertwig’s epithelial root sheath cells migrate (based on principles of and developmental bio- into the periodontal ligament site; a few undergo logy) for fabricating new tissues to replace or regen- apoptosis and some remain in the root surface erate lost tissues (205), it is important to understand (108). the formation of specific tissues, the physico-chem- Although it is accepted that the Hertwig’s epithelial ical characteristics of the tissues and the molecular root sheath plays an important role in root develop- events leading to the normal function of the tissues. ment, the precise nature of its role remains contro- versial. In 1940, Schour & Massler suggested that the major function of the Hertwig’s epithelial root sheath Development of the periodontium was to induce and regulate root formation, including the size, shape and number of roots (244). Other The periodontium can be defined as Ôan intricate investigators suggested that the role of the Hertwig’s mosaic of cells and proteins that is primarily epithelial root sheath was to induce the differentiation

196 Regeneration of periodontal tissues: cementogenesis revisited

Fig. 1. Root development and periodontium formation. continues, and there is formation of the periodontium Histological sections of 7-day postnatal mouse mandibu- with cementum, periodontal ligament and bone. Am, lar molars showing the initial development of the root by ; C, cementum; D, dentin; Ds, dental sac; formation of the Hertwig’s epithelial root sheath. At the HERS, Hertwig’s epithelial root sheath; Od, ; 14-day postnatal time-point, apical migration of the roots PDL, periodontal ligament. of odontoblasts to form the root dentin (183, 182, 222, chymal transformation to become functional ce- 243, 251), or to differentiate dental sac cells into mentoblasts in charge of producing the acellular (181). The current notion states that cementum (251, 275). Hertwig’s epithelial root sheath cells produce the The gingival tissues appear to be derived from both basement membrane containing chemotactic pro- the and the developing germ (135). teins, which serve to direct the migration of prece- It has been suggested that the dental follicle (con- mentoblast cells (140, 141, 182, 235, 251) and to nective tissue surrounding the developing teeth) induce differentiation (191, 232, 234). gives rise to the fibroblasts forming the periodontal Amongst the basement membrane molecules are ligament as well as to the alveolar bone and several extracellular matrix proteins, growth factors, cementoblasts (45, 136, 186, 243), all of which have enamel proteins and adhesion molecules, such as a a common origin (34). Therefore, it is collagenous-like protein, known as cementum postulated that there are different types of attachment protein (CAP), which has chemotactic cementoblasts: those originating from the Hertwig’s potential capable of recruiting putative cementoblast epithelial root sheath via epithelial–mesenchymal precursors (11, 149, 156, 196, 275). In the second transformation and which form the acellular stage of cementogenesis (when the tooth reaches cementum; and those derived from the dental follicle, and cellular cementum is formed), the which form the cellular cementum (9, 19, 105, 251, proliferation of cells of the Hertwig’s epithelial root 275). It is also believed that progenitors for perio- sheath is considerably reduced, and some cells are dontal ligament, and cementoblast cells entrapped in the newly formed mineral where they adopt a paravascular location in the periodontal may influence phenotypic changes in the dental sac ligament, and these cells, which exhibit some fea- cells (252). It is also suggested that Hertwig’s epi- tures of stem cells, can regenerate functional tissues thelial root sheath cells undergo epithelial–mesen- when the need arises (150–153, 195). Periodontal

197 Zeichner-David ligament stem cells have recently been isolated from may negatively regulate root resorption and induce the human periodontium (162, 224, 225). acellular cementum formation (56). In addition, cells The Epithelial Rest of Malassez cells remain in the of the Epithelial Rest of Malassez may help in ce- periodontal ligament throughout life, suggesting that mentum repair because of their ability to activate they have important, although yet unknown, func- matrix proteins, such as , which are also tions, rather than just being leftover structures. Roles expressed during tooth development (76, 81). attributed to the Epithelial Rest of Malassez cells In summary, based on the information presented, it range from bad to good. The Epithelial Rest of appears that the developed or ÔadultÕ periodontium Malassez cells are held responsible for the formation retains its potential for repair/regeneration in the form of periodontal cysts and tumors as a result of peri- of cells of the Epithelial Rest of Malassez, progenitor apical inflammation associated with pulpal necrosis cells and stem cells, which can be induced to differ- (26, 57, 77, 176, 226, 242). It has also been suggested entiate into cementoblast, osteoblast or periodontal that Epithelial Rest of Malassez cells contribute to the ligament cells to regenerate periodontal tissues. formation of the periodontal pocket because of their continuum with the junctional (176, 238). Molecular factors involved in Some studies report the ability of Epithelial Rest of periodontal development Malassez cells to resorb bone and extracellular mat- rix, and thus implicate the cells in root resorption (15, It is well known that tooth development is regulated by 75, 122). On the other hand, it has also been sug- temporal- and spatial-restricted reciprocal epithelial– gested that the cells of the Epithelial Rest of Malassez mesenchymal interactions. A number of genes that may protect the root from resorption (259). The play a crucial role in tooth development have been finding of Epithelial Rest of Malassez cells being identified and include growth factors and their closely associated with neural endings suggests that receptors, such as transforming growth factor b-1 they have a role in the development of periodontal and )2, bone morphogenetic protein-2 and )4 ligament innervation (126). Studies performed with (BMP-2, )4), activins, fibroblast growth factor-4, )8 1-hydroxyethylidene-1,1-bisphosphonate, a drug that and )9 (FGF-4, )8, )9), hepatocyte growth factor, and interferes with homeostasis in the periodontal liga- midkine and transcription factors, such as the home- ment, showed a severe reduction in the width of the obox genes (Msx1, Msx2, Dlx1, Dlx2, Dlx3, Otlx2, periodontal ligament with the development of anky- Barx1), Pax genes (Pax9 and Pax6), and Lef1, Gli2/Gli3 losis, which was repaired after discontinuing the and Shh (40, 100, 192, 249, 274). It has been docu- administration of 1-hydroxyethylidene-1,1-bisphos- mented that growth factors are involved in establish- phonate (261). As the study did not detect a change in ing the presence, number, site, size or shape of teeth. the number of Epithelial Rest of Malassez cells post- The availability of knockout mice has provided critical treatment, it was suggested that cells of the Epithelial information on some growth factors that are deter- Rest of Malassez are unlikely to play an important minants of early tooth development. However, little part in the homeostasis of, and may not be a prere- information is currently available on the growth and quisite for, the repair and maintenance of the perio- transcription factors involved in the later stages of dontal ligament. On the other hand, the Epithelial tooth development, such as root development. Al- Rest of Malassez cells secrete hyaluronic acid, which though one can assume that the same epithelial– contributes to the formation of the loose connective mesenchymal interactions will take place between the tissue characteristics of the periodontal ligament Hertwig’s epithelial root sheath and the underlying (155). Cells of the Epithelial Rest of Malassez react to ÔrootÕ mesenchyme, and all or some of the same growth mechanical stress, like that associated with ortho- factors will be involved in root formation, these issues dontic tooth movement, by increasing their prolifer- have been only minimally addressed. Transforming ation rate and cell size (27), and thereby help to growth factor b-1 and its receptors (58, 59), and BMP-2, maintain the space between the periodontal bone )3 and )7 (249), have been identified in cemento- and cementum to avoid ankylosis (134). The in- blasts, periodontal ligament and alveolar bone, and creased activity of the Epithelial Rest of Malassez BMP-2, )4 and MSX-2 have been reported in the cells is consistent with their putative role on Hertwig’s epithelial root sheath (266). turnover in the periodontal ligament, which is growth factor-2 (143), receptors for epidermal growth accelerated during tooth movement (241), and during factor (42) and growth hormone (270) have been cementum repair in areas of root resorption (24). It is detected in periodontal tissues. However, the pub- suggested that the Epithelial Rest of Malassez cells lished studies are all descriptive and do not provide

198 Regeneration of periodontal tissues: cementogenesis revisited information as to the function of these growth factors cementum. As in bone and dentin, the major com- in periodontium development. Furthermore, the ponent of cementum is collagen (16). The expression transforming growth factor-b1-knockout mouse dis- of noncollagenous proteins that stimulate cell migra- plays no apparent defects in tooth and root develop- tion, attachment, proliferation, protein synthesis and ment (39), thus excluding a role for this factor in these mineralization during root formation has been processes. On the other hand, by using transgenic mice reported by several investigators (38, 142, 147). In the that express the BMP inhibitor, noggin, driven by the early stages of root development, immunohisto- keratin 14 promoter (K14-noggin), we recently dem- chemical techniques have shown the expression of onstrated that BMPs are important for proper root multifunctional proteins, such as laminin and morphogenesis. When the function of BMPs is re- fibronectin (140). These proteins, as well as other pressed, the transgenic mice demonstrate a delay in proteins extracted from cementum (173), are initially tooth development, lack of enamel formation and believed to function as chemo-attractants. Laminin abnormally shaped roots (198). Insulin-like growth and fibronectin can also function as adhesion pro- factor-I receptor has been demonstrated in the Her- teins, together with tenascin (137), bone sialoprotein twig’s epithelial root sheath, and in vitro experiments (38, 142), osteopontin (25), and a 55-kDa cementum- suggest that insulin-like growth factor-I receptor plays attachment protein (196, 263). The presence of other a role in the proliferation and elongation of the Her- bioactive proteins, such as enamel-like proteins (235, twig’s epithelial root sheath, which is critical for root 234), osteonectin/SPARC (201), and mitogenic factors development (55). (157, 269), have also been reported in the cementum. Transcription factors associated with root develop- In addition to these proteins, cementoblasts synthes- ment include two members of the homeobox family of ize and secrete several glycosaminoglycans (such as transcription factors: Dlx2 and Dlx3. The expression of chondroitin-4-sulfate, chondroitin-6-sulfate and der- Dlx2 by the Hertwig’s epithelial root sheath during root matan sulfate, and collagen fibrils), which are present development was demonstrated using Dlx2/LacZ in the cemento–dentinal junction (88, 264, 265). transgenic mice (132). Although these studies are only It has been suggested that cementoblasts exhibit suggestive of a role of Dlx2 in root development, it was an osteoblast-like, rather than an - of interest that the Dlx2 knockout mice showed normal like, phenotype (25). Odontoblast, osteoblast and teeth, while the Dlx1/Dlx2 knockout mice lacked cementoblast cells express several matrix proteins, maxillary molars (253). The involvement of Dlx3 in root such as osteopontin, bone sialoprotein (BSP), osteo- development comes from the phenotype expressed by nectin, osteocalcin, matrix Gla protein (208) and den- patients affected with the genetic disease, tricho- tin-matrix-protein 1 (DMP-1) (106). The presence of dento-osseous syndrome, which presents root defects osteocalcin in cementum is more controversial. as well as defects in hair, bone and enamel. A deletion Bronckers et al. (25), using immunohistochemistry, of 4 bp in the Dlx3 gene, which causes a frameshift reported the presence of osteocalcin on the cellular mutation and premature codon termination, resulting intrinsic fiber cementum (CIFC) and associated in an altered protein, were identified in a family with cementoblasts (mature), but not in the acellular tricho-dento-osseous syndrome (199). We recently cementum and its associated cementoblasts. Tenorio reported the importance of the Nfi-c transcription et al. (246) reported the presence of osteocalcin in factor in root development. Nfi-c knockout mice ap- acellular extrinsic fiber cementum (AEFC) but not in pear normal, except that they exfoliate their teeth the associated cementoblasts, while CIFC and associ- shortly after eruption. These mice show a lack of roots ated cementoblasts stained weakly. Bosshardt & Nanci of both mandibular and maxillary teeth, and therefore (20) used two different antibodies (OC1 and OC2), their teeth have no bone attachment. Histological which gave different results: OC1 showed reactivity analysis indicated a normal crown, enamel and dentin with acellular cementum, while OC2 was negative. formation, and although there is initial formation of Similarly, the presence of DMP-1 has been associated the Hertwig’s epithelial root sheath and a budding with acellular cementum (275) and cementocytes, but root, no further development occurs of the roots, ce- not with cementoblasts (255). It has been suggested mentum and periodontal attachment apparatus (239). that acellular cementum is a unique tissue, while cel- lular cementum and bone share some similarities, Cementum composition although there are still morphological, functional and biochemical differences between the two tissues (19). In order to understand the process of cementogenesis, The presence of cementum-specific proteins it is important to determine the composition of remains questionable, although some putative

199 Zeichner-David cementum-specific proteins have been invoked: a periodontal ligament cells are removed from the ce- 55-kDa CAP (263); a mitogenic factor (167); and a mentum or are unable to regenerate, bone tissue in- 72-kDa protein, CEM-1 (235). However, as the char- vades the periodontal ligament space and establishes acterization and the sole expression by cementoblasts a direct connection between the tooth and the wall of of these proteins have not been determined, the the alveolar socket, resulting in ankylosis. The ankyl- possible existence of cementum-specific proteins otic, nonflexible type of tooth support can lead to loss remains unknown. It has been reported that of function and resorption of the tooth root (13). cementoblasts and cementocytes produce high levels of the GLUT-1 monosaccharide transporter, while Can guided tissue regeneration and bone or do not express this protein. grafting regenerate cementum? These data suggest that GLUT-1 may play a role in cementogenesis and could serve as a biomarker to Nyman et al. (174), using Millipore membranes, differentiate between cells of cementoblastic and introduced the concept of a membrane barrier, which osteoblastic lineage (124). However, the observed excludes the apical migration of gingival epithelial differences in GLUT-1 are quantitative, and GLUT-1 is cells and provides an isolated space for the inwards present in many different cell types. Recently, we migration of periodontal ligament cells, osteoblasts reported the isolation of a -derived and cementoblasts. Guided tissue regeneration was protein, CP-23, that is expressed by cementoblasts and successfully used to aid in the regeneration of lost some precursor cells present in the periodontal liga- periodontal tissues caused by periodontitis (67). The ment, but not by osteoblasts. The function of the CP-23 first guided tissue regeneration membranes were protein is currently unknown; however, given its nonabsorbable and made of polytetrafluoroethylene, nuclear location, it may be required for cementoblast such as Gore-Tex. Studies on experimentally differentiation and may be used as a marker for induced periodontal defects in monkeys suggested cementoblast cells (3). The CP-23 protein is also ex- that guided tissue regeneration was capable of pressed by Hertwig’s epithelial root sheath cells (275). inducing the formation of new bone and cementum Based on our current knowledge of the develop- (4). The second generation of guided tissue regener- ment of periodontal tissues, several strategies exist ation used absorbable membranes made of collagen for targeting regenerative therapy, ranging from or polylactic and citric acid (28, 159), which elimin- inducing their own ÔregenerativeÕ mechanisms using ated the need for surgical membrane retrieval (66). molecular approaches, or utilizing cells to repopulate Recent systematic reviews indicate that, in the and recapitulate the developmental process. treatment of intrabony and furcation defects, guided tissue regeneration is more effective than open flap . Various barrier types yielded no sys- Strategies for periodontal tematic difference in clinical outcome, but barrier regeneration/repair types could explain some heterogeneity in the results. Overall, guided tissue regeneration is consistently The process of periodontal tissue regeneration starts more effective than open flap debridement in the at the moment of tissue damage by means of growth gain of clinical attachment and reduction of probing factors and cytokines released by the damaged con- depth in the treatment of intrabony and furcation nective tissue and inflammatory cells. It is well defects (99, 163). The use of grafting material in accepted that in order to improve periodontal healing, combination with guided tissue regeneration seems root planing or root conditioning is a necessary to improve clinical outcomes for furcation, but not antecedent to mesenchymal cell migration and for intrabony defects, when compared with the use of attachment onto the exposed root surface. Acid barrier membranes alone. It has also been questioned treatment, in particular with citric acid, has been whether guided tissue regeneration produces true found to widen the orifices of dentinal tubules, cementum regeneration or only cemental repair. The thereby accelerating cementogenesis and increasing newly formed cementum has been characterized as a cementum apposition and connective tissue attach- cellular cementum that is usually poorly attached to ment. However, a systematic review performed by the dentin surface (125). It is suggested that perio- Mariotti (145) suggested that the use of citric acid, dontal healing with guided tissue regeneration ther- or EDTA to modify the root surface pro- apy occurs in two stages. The first stage comprises an vides no clinical significant benefit for regeneration in initial healing phase with the formation of a blood patients with . Conversely, when clot, transient root resorption/demineralization,

200 Regeneration of periodontal tissues: cementogenesis revisited deposition of acellular cementum on the root surface regulate tooth development and tissue repair, sug- and formation of connective tissue. The second gests the use of some of these factors for periodon- phase comprises a remodeling process, which will tium regeneration (37, 61, 68, 71, 118, 116, 117, 128, result in a regenerated cementum similar to pristine 170). Some attachment proteins, such as fibronectin cementum as maturation proceeds over time (69). In (29, 206, 262) or CAP (156, 196), are able to enhance conclusion, several clinical studies have demonstra- fibroblast migration, attachment and orientation of ted that guided tissue regeneration is a successful the connective tissue to the root surface. New treatment modality for periodontal reconstructive strategies, utilizing growth factors to induce cell surgery and it has become an accepted procedure in migration, proliferation and differentiation, were most periodontal practices, either by itself or in developed to repopulate the damaged periodontal combination with other treatment modalities. tissues with periodontal ligament cells (32, 247). It is Autologous bone grafts to repair periodontal osse- believed that growth factors play important roles in ous defects have been used for many years and dif- modulating the proliferation and/or migration and/ ferent approaches have been the subject of several or differentiation of structural cells in the periodon- reviews (165, 209). Bone repair can also be achieved tium (58, 86, 97, 197, 230). It is suggested that growth using ceramic materials such as Bioglass, which is a factor molecules are produced during cementum bone-bonding bioactive material that has been widely formation and then stored in the mature cementum used for (110). Studies in monkeys sug- matrix with the potential to induce periodontal repair gested that PerioGlas (synthetic bone particulate) or regeneration when needed (236). Large-scale pro- can achieve superior bone repair and cementum duction of recombinant growth factors has facilitated regeneration and retard epithelial down-growth in vitro and in vivo studies to determine the efficacy compared with other, similar materials (50, 109). of growth factors in periodontal tissue regeneration. Additionally, these materials can be used as scaffolds Amongst the growth factors currently being used or to deliver other bioactive molecules to enhance their are platelet-derived growth factor, insulin-like growth function. The use of bone grafts, powders or ceramics factor (36, 63, 92, 138, 188, 210), transforming growth is quite prevalent in many dental practices. A recent factor-b1 (146), basic fibroblast growth factor (213), systematic review on the efficacy of bone replacement dexamethasone (211) and BMPs (121, 205, 211). grafts compared with other interventions in the treat- However, problems in applying these growth factors ment of periodontal osseous defects was performed by for periodontal repair include the nonspecific activity Reynolds et al. (202). Meta-analysis indicated that for of some factors on different cell lineages in time and the treatment of intrabony defects, bone grafts are space, and the rapid loss of growth factors applied effective in reducing crestal bone loss, increasing bone topically (13, 138). level, increasing clinical attachment level, and redu- It has been shown that both platelet-derived cing probing depth compared with open flap debri- growth factor and insulin-like growth factor-1 can dement procedures. Histological studies showed that stimulate the proliferation and chemotaxis of perio- demineralized freeze-dried bone allografts support the dontal ligament cells, and that the combination of formation of a new attachment apparatus in intrabony platelet-derived growth factor and insulin-like growth defects; however, the available data indicate that factor-1 can further increase the mitogenic effect (23, alloplastic grafts support periodontal repair rather 44, 175). In addition to the mitogenic activity, plate- than regeneration, and that the best treatment is a let-derived growth factor also appears to stimulate combination of bone grafts with barrier membranes. collagen synthesis in periodontal ligament cells (146). Nevertheless, these strategies are directed mainly to Furthermore, dexamethasone has been shown to enhance alveolar bone and periodontal ligament exert the same effect as insulin-like growth factor-1 repair and have the problems that they do not address on periodontal ligament fibroblasts, gingival fibro- cementogenesis and therefore do not completely blasts and fibroblasts, and may substitute for regenerate the architecture of the original periodon- insulin-like growth factor-1 in the platelet-derived tium. growth factor stimulation of cell proliferation (210). In addition to the previously described effects, Molecular approaches for cementum platelet-derived growth factor has the capacity to regeneration significantly negate and reverse the inhibitory effects of lipopolysaccharide on the proliferation of human Advances in our knowledge of developmental bio- gingival fibroblasts. Lipopolysaccharide from a vari- logy, and of the growth factors that initiate and ety of gram-negative bacteria is known to inhibit

201 Zeichner-David gingival fibroblast proliferation and synthesizing biological activity (7, 6, 65). The potential use of gene activity, has been implicated in periodontal inflam- therapy in vivo to stimulate periodontal tissue mation and may also be responsible for delayed regeneration has been studied in large tooth-associ- wound healing following periodontal therapy (12). ated alveolar bony defects in rats. The results showed In vivo studies using the beagle dog (natural perio- that the direct gene transfer of platelet-derived dontal disease) and the nonhuman primate (ligature- growth factor-B stimulates the regeneration of induced attachment loss) models showed that the alveolar bone and cementum (104). application of platelet-derived growth factor/insulin- As stated above, some members of the BMPs are like growth factor-1 resulted in significant amounts of normally expressed during the development of the new bone and cementum formation (138, 210). periodontium, such as BMP-3 and BMP-7/OP-1, Treatment with insulin-like growth factor-1 alone did which have been localized immunologically in not significantly alter healing compared with controls, alveolar bone, cementum, and periodontal ligament, while treatment with platelet-derived growth factor whereas BMP-2 was only localized in the alveolar alone showed significant regeneration of attachment. bone (249, 266). Although the exact role of BMPs in Although there are differences in the response to the development of the periodontium has not yet platelet-derived growth factor/insulin-like growth been determined, these proteins are good candidates factor-1, depending on which animal model is used for stimulating periodontal regeneration because of (the osseous response in dogs appears to be greater their ability to promote not only osteogenesis but than that of the nonhuman primate, while new also cementogenesis. The expected role of BMPs in attachment formation appears to be greater in the stimulating intramembranous bone formation with- nonhuman primate than in the dog), there is consis- out an endochondral intermediate may provide tency in promoting periodontal regeneration (63, 64). greater osteogenic potential than autogenous bone or Rutherford et al. (211) showed that platelet-derived other bone substitutes (121, 118, 119, 170, 205, 240). growth factor and dexamethasone, combined with a Studies indicate that recombinant BMP-2 exerts no collagen carrier matrix, induced regeneration of the effect on the growth and differentiation of human periodontium in monkeys. It has also been shown that periodontal ligament cells in vitro; however, BMP-2 the combination of platelet-derived growth factor and stimulates alkaline phosphatase activity and para- guided tissue regeneration work better than either of thyroid hormone-dependent 3¢,5¢-cyclic adenosine the two modalities alone (36, 188). monophosphate (cAMP) accumulation, which are Clinical trials in humans using platelet-derived early markers of osteoblast differentiation. Never- growth factor/insulin-like growth factor to treat per- theless, BMP-2 produced no mature osteoblasts, as iodontal osseous defects showed that only high doses measured by expression of osteocalcin, and also of these factors gave rise to a statistically significant inhibited 1,25(OH)2D3-induced osteocalcin synthesis increase in alveolar bone formation (92). When in these cells (123). In vitro studies using mouse-de- platelet-derived growth factor was used in combina- rived dental follicle and periodontal ligament cells tion with bone allografts to treat Class II furcations suggest that BMP-2 induced dental follicle cells to and interproximal intrabony defects, histological differentiate towards a cementoblast/osteoblast phe- evaluation showed regeneration of new alveolar notype but had no effect on periodontal ligament cells bone, cementum, and periodontal ligament (30, 171). (278). Paradoxally, BMP-2 was found to inhibit ce- Platelet-rich plasma is a fraction of plasma that mentoblast cell mineralization in vitro by decreasing contains platelet-derived growth factor and trans- the expression of BSP and collagen type 1 (279). In forming growth factor-b (180). An alternative to the studies of BMP-2 on early wound healing in a rat use of recombinant growth factors is the use of a model of periodontal regeneration, the connective platelet gel in combination with demineralized tissue attachment was found to be similar in animals freeze-dried bone allografts (5, 43). receiving BMP-2 and in controls. However, BMP-2 The limitations of topical protein delivery to peri- induced bone formation at some distance from the odontal osseous defects include transient biological defect, which indicates the need for a suitable delivery activity and bioavailability of platelet-derived growth system to maintain the BMP-2 at the site of implan- factor at the wound site. To overcome these limita- tation (120). Other studies suggest that the effects of tions, studies have used genetic engineering to BMPs may be influenced by certain factors, such as transduce cells derived from the periodontium, using root surface conditioning, delivery systems, mastica- adenovirus carrying the platelet-derived growth fac- tory forces, etc., and that BMP-2 stimulates the pro- tor gene to promote sustained release and ensure liferation and migration of cells from the adjacent

202 Regeneration of periodontal tissues: cementogenesis revisited periodontal ligament into the wounded area, pro- keys, followed by histological analysis that showed moting new cementum formation (119). almost complete regeneration of acellular cementum, The expression of both BMP-2 and BMP-7 during firmly attached to the dentin and with collagenous periodontal tissue morphogenesis suggests that fibers extending towards newly formed alveolar bone optimal therapeutic regeneration may require the (79). These studies resulted in a new therapeutic combined use of the two BMPs. BMP-7-treated molar preparation to treat periodontal disease, consisting of furcation defects in baboons resulted in substantial hydrophobic enamel matrix proteins extracted from cementogenesis, while BMP-2 showed limited porcine developing enamel, which has been marke- cementum formation but greater amounts of miner- ted by Biora, Inc., under the name of Emdogain.In alized bone and osteoid; however, the combined the past 8 years, the use of enamel proteins for application did not enhance alveolar bone regener- inducing the formation of cementum, bone and ation or new attachment formation over and above dentin has generated numerous in vivo and in vitro that obtained by separate applications of the two studies, as well as clinical trials, resulting in almost BMPs (207). Recently, it was shown that the appli- 300 publications. In vitro studies, animal studies and cation of a synthetic BMP-6 polypeptide to a perio- clinical trials are all being conducted simultaneously dontal fenestration defect in rats resulted in (60, 70, 83, 154). increased formation of new bone and cementum In vitro studies, using periodontal-associated cells (93). Perhaps the use of other members of the BMP such as periodontal ligament fibroblasts, osteoblasts, family, such as growth and differentiation factor-5, cementoblasts, gingival fibroblasts, gingival epithelial )6, and )7, might provide better and more complete cells, etc., have been conducted in an attempt to regenerative outcomes. These factors have been understand the molecular and cellular mechanisms detected during the process of periodontal develop- involved in the process of - ment at the surfaces of alveolar bone, cementum and induced tissue regeneration. In order for enamel periodontal ligament fiber bundles (223). matrix derivative to regenerate periodontal tissues, it Limitations for the regular use of BMPs are the will need to exert an effect on proliferation, migra- need for high doses, non-specific activity on different tion, attachment and/or differentiation of the sur- cell lineages in time and space, and the rapid loss of rounding periodontal cells, and most studies have topically applied growth factors (13, 138). Some of measured these parameters, as shown in Table 1. these problems can be overcome by the use of gene Few studies have tested the effect of enamel matrix transfer technology. Jin et al. (103) used adenoviruses derivative on cell migration, but available data sug- containing BMP-7 to transduce dermal fibroblasts gest an increased migration of periodontal ligament that were then used to treat mandibular alveolar cells, osteoblasts, gingival fibroblasts and dermal bone defects in a rat wound repair model. Their fibroblasts in response to enamel matrix derivative, results showed chrondrogenesis, with subsequent with the exception of one study that found no effect osteogenesis, cementogenesis and bridging of the on periodontal ligament cells (184). Most studies on periodontal bone defects, suggesting that this genetic the effect of enamel matrix derivative on cell engineering approach may be useful in alveolar bone attachment, which generally included periodontal regeneration. A recent literature review (62) conclu- ligament cells, found an increase in cell attachment ded that although promising, there were insufficient (184). However, one study found the enamel matrix data at the present time to conduct a meta-analysis derivative to have no effect on cell attachment of on the effect of growth factors for periodontal repair, gingival fibroblasts (256). A number of studies, which and pointed to the need for more clinical trials. measured the effect of enamel matrix derivative on cell proliferation, have found an increase in cell Do enamel-associated proteins proliferation in the presence of enamel matrix regenerate cementum? derivative. However, the proliferative effect was not found in two studies using periodontal ligament cells Based on the presence of enamel proteins in acellular (41, 256), in two studies using osteoblast cell lines cementum (133, 235, 182, 233), it was thought that (215, 268) and in one study using gingival fibroblasts these proteins may play a role in the repair/regen- (256). Several studies found an inhibition of cell eration of periodontal tissues destroyed by perio- proliferation when epithelial cells were used (112, dontal disease (78). This idea was tested by adding 139, 273). These data may explain the clinical enamel proteins or purified enamel matrix derivative observation that application of enamel matrix to surgically produced periodontal defects in mon- derivative suppresses the down-growth of junctional

203 Zeichner-David 204 Table 1. In vitro studies on the effect of enamel protein derivative on cells Cells Species Migration Attachment Proliferation Differentiation Mineralization Reference Periodontal ligament cells Human ND ND ND AP increase – osteoblast Yes (166) Human No effect No effect Yes – increase No (Type I col) ND (184) Rat (primary) ND ND Yes – decrease No – Col, AP (95) Human (primary) ND ND Yes – increase Yes – less AP – cementoblast ND (33) Human (primary) Yes ND Yes – increase ND ND (203) Human (primary) ND Yes No difference ND ND (41) Human (primary) ND ND Yes – increase Yes, increase IGF-1 and TGF-b1. ND (178) No effect on bone phenotype Hu (primary) ND ND ND Increase matrix (versican, biglycan, ND (73) decorin, hyaluronan Hu (primary) ND Yes No effect Increase AP and TGF-b1 ND (256) Hu (primary) Yes ND Yes – increase ND ND (89) Hu (primary) ND Yes Yes – increase Increase cAMP, TGF-b1, IL-6, PDGF-AB ND (139) P (primary) ND Yes Yes – increase Increase OPN ND (204) Mo (cell line)** ND Yes Yes – increase Inhibits Col I, de novo expression BSP ND (273) and OCN, increase BMP2 Mo (cell line) ND Yes Yes – increase Inhibits Col I, de novo OCN and BMP3 ND (273) Osteoblasts Hu (ROS17/2.8) ND ND ND BSP increase ND (227) Hu (primary) ND ND Yes – increase More FGF2 and COX2; less AP and MMP1 ND (161) Mo (ST2) ND ND No effect Yes – AP ND (268) Mo (KUSA/A1) ND ND Yes – increase Yes – AP, Col, OPN, TGF-b1, OCN Yes- more (268) and MMPS Mo (primary) ND ND Yes – increase ND ND (101) Mo (primary) ND ND ND Increase Col, IL-6 and PGHS-2; ND (102) no effect on OCN and IGF-1 Mo (MC3T3-E1) ND ND Yes – increase Increase OPN and less OCN (254) Hu (2T9 pre-osteoblasts) ND ND Yes – increase No effect ND (215) Hu (MG63 osteoblast like) ND ND Yes – decrease Yes, increase AP, OCN, TGB1 ND (215) Table 1. Continued

Cells Species Migration Attachment Proliferation Differentiation Mineralization Reference Hu (primary) ND ND Yes – increase Yes, increase AP, OCN, TGB1 ND (215) Hu (MG63) Yes ND Yes – increase ND ND (89) P (primary) ND Yes Yes – increase Increase OPN ND (204) Hu (Ros17/28)* ND ND ND BSP increase ND (228) Gingival fibroblast cells Rat ND ND Yes – double Faster – osteogenic Yes – more (115) Hu (primary) Yes ND Yes – increase ND ND (203) Hu (primary) ND ND ND Increase matrix (versican, biglycan, ND (73) decorin, hyaluronan Hu (primary) ND No effect No effect Increase AP and TGF-b1 ND (256) Hu (primary) Yes ND Yes – increase ND ND (89) Rat ND ND Yes – increase More ECM No (115) Rat ND ND No difference ND ND (72) P (primary) ND Yes Yes – increase Increase OPN ND (204)

Dental follicle Mo (SV40) ND ND Yes – increase More OPN, Less OCN Inhibits (74) revisited cementogenesis tissues: periodontal of Regeneration Cementoblasts Mo (SV40) ND ND Yes – increase Decrease Ocn Inhibits (254) Mo (OCCM-30)* ND ND ND Decrease BSP Inhibits (258) Mo (OCCM-30)NDà ND No effect Decrease OCN, increase OPN and OPG Inhibits (17) Mo (L929) ND ND No difference ND ND (72) Rabbit Yes – vascular endothelium. Growth factors (160) Human (primary) Yes ND Yes – increase ND ND (203) Mesenchymal stem cells Hu (C2C12) ND ND ND Yes – increase AP. Osteoblast phenotype ND (177) Epithelial cells Hu (HELA) ND ND Inhibited Increase cAMP and PDGF-AB ND (113) Hu (SCC25) ND ND Inhibited Increase p21WAF1/cip1; decrease CK-18 ND (113, 112, 114) ERM P (primary) ND Yes Yes – increase Increase OPN ND (204) Endothelial cells Hu (HUVEC) Yes – increase ND No effect ND ND (271)

AP, alkaline phosphatase; BMP, bone matrix protein; BSP, bone sialoprotein; Col, collagen; Hu, human; IGF-1, insulin-like growth factor; IL-6, interleukin-6; MMPS, matrix metalloproteinases; Mo, mouse; ND, not determined; 205 OCN, osteocalcin; OPN, osteopontin; OPG, osteoprotegerin; P, pig; PDGF-AB, platelet derived growth factor AB; PGHS-2, prostaglandin G/H synthase 2; TGF-b1, transforming growth factor b-1. *Mouse recombinant amelogenin. Mouse recombinant . àMouse leucine rich amelogenin peptide (LRAP). Zeichner-David epithelium onto dental root surfaces, a process that that BMPs are the molecules responsible for enamel frequently interferes with the formation of new con- matrix derivative activity. Although these studies nective tissue attachments (79, 78). suggest that the action of Emdogain is a result of The majority of available in vitro studies have the presence of contaminating growth factors, other analyzed the effect of enamel matrix derivative on studies have shown that pure recombinant enamel gene expression and differentiation, and most of proteins indeed have activity as inducers. The results these studies found either an increased or a de- obtained in our laboratory indicate that mouse creased expression of certain transcription and recombinant amelogenin can increase attachment growth factors, extracellular matrix proteins or min- and proliferation of mouse periodontal ligament cells eralization-associated proteins in the cells tested. in vitro (272, 273). Furthermore, a post-translational Where mineralization was measured, it was found modified recombinant ameloblastin, another enamel- that enamel matrix derivative induced mineralization associated protein, had an effect similar to that of of periodontal ligament cells (166), increased miner- amelogenin on periodontal ligament cells. Both alization of osteoblasts (268) and gingival fibroblasts recombinant amelogenin and ameloblastin can (115), decreased mineralization of cementoblast cells change the phenotype expressed by periodontal (254) and inhibited the mineralization of dental fol- ligament cells by inhibiting the expression of colla- licle cells (74). Differences in results amongst studies gen type I and inducing de novo expression of can be explained by differences in sources and con- osteocalcin. Amelogenin also induced the expression centrations of enamel matrix derivative and in the of bone sialoprotein and BMP-2, while ameloblastin cell preparations used. Most studies employed pri- induced the de novo expression of BMP-3 (273). mary cell cultures derived from different patients, These results indicate that both enamel-associated which probably contained mixed populations of a proteins have a modulatory effect on the expression variety of cells present in the periodontium. Never- of BMPs, suggesting that perhaps these proteins exert theless, taken together, these studies suggest that their signaling effect by means of BMPs. Recombin- enamel matrix derivative can act as a multipurpose ant mouse amelogenin improved osteoblast adhe- growth factor capable of stimulating the proliferation sion (90), and increased the expression of bone of mesenchymal cells while inhibiting the cell divi- sialoprotein and decreased the formation of miner- sion of epithelial cells, and can stimulate attachment alized nodules in cementoblasts (258). A leucine-rich and phenotypical changes in some cells, while amelogenin peptide, which exhibited no effect on inhibiting matrix production in others. cell proliferation, down-regulated osteocalcin and Given the widespread use of Emdogain, and the up-regulated osteopontin in a dose- and time- fact that it is made from an extract of enamel pro- dependent manner, and inhibited the capacity to teins, it is important to identify the actual protein form mineral nodules (17). Taken together, these responsible for its function. Studies by Maycock et al. reports point towards a growth factor activity for (148) found that, in addition to amelogenin, Emdo- enamel proteins that may be of importance in gain contains metalloproteases and serine pro- periodontal tissue regeneration. teases. Studies by Kawase et al. (114) demonstrated Several clinical trials have shown an increase in that porcine enamel matrix derivative contains periodontal attachment and bone formation in indi- transforming growth factor-b1 (or a transforming viduals treated with Emdogain (54, 85, 87, 154, 179, growth factor-b-like substance), and that the action 200, 217, 216, 218, 219, 277). However, in many of of enamel matrix derivative is mediated by the smad- these studies, the results were no better than those 2 signaling pathway. In addition, a neutralizing obtained with other previously established treat- anti-transforming growth factor-b immunoglobulin ments, such as guided tissue regeneration, which blocked the action of enamel matrix derivative on yields better outcomes in the management of deep epithelial cells, although it failed to block completely intrabony periodontal defects (84, 187, 218, 221, 231). enamel matrix derivative-induced fibroblastic prolif- Histological studies revealed that treatment with eration, suggesting the presence of more than one Emdogain is unpredictable, resulting in the forma- growth factor. Iwata et al. (98) isolated the inductive tion of cellular cementum rather than acellular activity of enamel matrix derivative by using chro- cementum, and this cementum was only partially matography and characterized it as being BMP-2 and attached to the root surface, similar to the cementum BMP-4 using specific antibodies. Furthermore, in the formed with the use of guided tissue regeneration. presence of noggin (an inhibitor of BMPs), enamel Furthermore, more bone regeneration occurred by matrix derivative lost its inductive activity, indicating using a guided tissue regeneration procedure than

206 Regeneration of periodontal tissues: cementogenesis revisited

Emdogain (216, 219, 218). Other studies showed no tum and promotes attachment of the supporting evidence of improvement in radiographic bone level, periodontal tissues, human histological studies have and surgical re-entry found new tissue with a rubbery questioned both the consistency of the histological consistency and that was not mineralized (189, 190). outcomes and the ability of enamel matrix derivative Experiments in rats, using a wounded rat periodon- to predictably stimulate the formation of acellular tium model followed by immunohistochemical cementum (107). It appears that following treatment analysis, showed that Emdogain does not affect the with enamel matrix derivative, a bone-like tissue expression of differentiation markers or bone matrix resembling cellular intrinsic fibrous cementum is protein synthesis in the repopulation response of formed (22). wounded rat molar periodontium (35). Despite the mixed results obtained from both Systematic studies, using literature reviews and in vitro and in vivo studies, new applications of meta-analysis, suggest that treatment with enamel Emdogain are continuously being reported. Some matrix derivative results in significant variations in studies suggest that it has the ability to induce the clinical outcomes (107). Although Emdogain is able formation of reparative dentin in pulpotomized teeth to significantly improve probing attachment levels (94, 96, 168, 169). It is being used to coat titanium and pocket depth reduction, some studies found no implants with mixed results; one study suggests that evidence of clinically important differences between there is enhanced formation of trabecular bone (229) guided tissue regeneration and Emdogain (47, 62) while the other found no effect (53). It has also been and reported that guided tissue regeneration is more suggested that enamel matrix derivative can combat predictable for cementum and bone regeneration bacteria in postsurgical periodontal wounds, which (257). Although animal histological studies with sur- otherwise could hamper wound healing and reduce gically created defects suggest that enamel matrix the outcome of regenerative procedures (8, 172, 220, derivative induces the formation of acellular cemen- 237). More recently, an acceleration of skin wound

PLF PL-7 DPM ControlHERS-CM Control HERS-CM Control HERS-CM

14 days

21 days

28 days

35 days

Fig. 2. Effect of Hertwig’s epithelial root sheath-condi- mentoblasts (PL-7) and dental papillae mesenchyme tioned media (HERS-CM) on periodontium-associated cell fibroblasts (DPM) were prepared from Immortomouse mineralization. HERS-CM was prepared by growing the (275). Cells were grown in differentiation conditions cells in Dulbecco’s modified Eagle’s minimal essential (DMEM supplemented with 10% FCS, 100 U/ml of peni- medium (DMEM) supplemented with 10% fetal calf serum cillin/streptomycin, 50 mg/ml of ascorbic acid and 2 mM (FCS) and 100 U/ml of penicillin/streptomycin. Cells were sodium b-glycerophosphate), with or without (controls) incubated at 39.5C in a humidified atmosphere of 95% 100 lg of HERS-CM proteins. At different time-points of air and 5% CO2 for 7 days, after which the media were culture, cells were fixed with 70% methanol and 30% collected, the protein concentration determined and then acetic acid and stained with Von Kossa to determine lyophilized. Periodontal ligament fibroblasts (PLF), ce- mineralization.

207 Zeichner-David healing in the presence of enamel matrix derivative the Epithelial Rest of Malassez cells, we are exploring was reported (160). the ability of these cells, or their secreted products, to induce periodontal ligament cells to differentiate into Cellular tissue engineering for cementoblasts in vitro. When periodontal ligament cementum regeneration cells, which do not produce a mineralized extracel- lular matrix, are grown in the presence of Hertwig’s It has long been recognized that a recolonization of epithelial root sheath conditioned media (HERS-CM), periodontal ligament cells onto the root surface is these cells produce a mineralized extracellular mat- necessary for periodontal ligament regeneration (129, rix, as determined by a positive Von-Kossa staining 174). One therapeutic approach proposed the removal of autologous cells from the patient’s periodontal Effect of HERS-CM on PLF ligament, culture of the cells in vitro, to place them cell differentiation back onto the exposed root coated with chemo- attractant factors, and then to cover the area with an P 21d 21d + HERS artificial basement membrane (247). A pilot study was carried out with four patients, using hydroxyapatite as BSP a vehicle for cell delivery. After 6 months, the treated patients exhibited greater pocket reduction and clin- ical attachment gain, and less , than OCN control patients; however, both groups showed good fill of the osseous defects studied (48, 49, 91). OSN Lekic et al. (130) tracked the fate and differenti- ation of rat periodontal cells and bone marrow cells transplanted into periodontal wounds in rats using OPN cells constitutively expressing b-galactosidase as a marker. Labeled cells were localized in the perio- dontal ligament and regenerating alveolar bone and it AP was suggested that, following a cyclical process of growth and development, both cell types were able to differentiate into periodontal ligament fibroblasts, BMP4 osteoblasts and cementoblasts, and to contribute to periodontal regeneration (131). Regeneration of cementum, periodontal ligament and alveolar bone Col1 has also been observed using auto-transplantation of bone marrow mesenchymal stem cells into perio- Actin dontal osseous defects in dogs (111). Similar results have been observed after the application of perio- dontal ligament cell sheets (2). Fig. 3. Effect of Hertwig’s epithelial root sheath-condi- The ability of cementoblasts and dental follicle tioned media (HERS-CM) on the phenotype of periodontal cells to promote periodontal regeneration in a rodent ligament cells. HERS-CM was prepared as previously described. Periodontal ligament cells were grown under periodontal fenestration model was analyzed recently proliferation (P) conditions (in the presence of interferon- (280). The results indicated that cementoblast-trea- c at 33C) or differentiation conditions [Dulbecco’s ted and carrier alone-treated defects showed com- modified Eagle’s minimal essential medium (DMEM) plete bone bridging and periodontal ligament supplemented with 10% fetal calf serum (FCS), 100 U/ml formation; however, no new cementum was formed of penicillin/streptomycin, 50 mg/ml of ascorbic acid and 2 mM sodium b-glycerophosphate] with or without (con- along the root surface in either group. Puzzling, trols) 100 lg of HERS-CM proteins. Cells were collected however, was the fact that no repair, or even osteo- after 21 days in culture (media were changed every other genesis, was seen within dental follicle cell-treated day), the media were removed, cells were rinsed in defects, even though these cells are believed to be phosphate-buffered saline (PBS) and total RNA was precursors of cementoblasts and to be responsible for extracted for determination of phenotype by using reverse transcription–polymerase chain reaction (RT–PCR). AP, alveolar bone formation. alkaline phosphatase; BMP-4, bone morphogenetic pro- As our laboratory has established immortal cell tein-4; BSP, bone sialoprotein; Col1, collagen type I; OCN, lines for the Hertwig’s epithelial root sheath (275) and osteocalcin; OPN, osteopontin; OSN, osteonectin.

208 Regeneration of periodontal tissues: cementogenesis revisited

(Fig. 2). This effect is specific for periodontal liga- References ment cells because other types of fibroblasts, such as those derived from the dental pulp, do not produce a 1. Adams DF. Diagnosis and treatment of refractory perio- mineralized extracellular matrix, even in the presence dontitis. Curr Opin Dent 1992: 2: 33–38. of HERS-CM. When cementoblast cells, capable of 2. Akizuki T, Oda S, Komaki M, Tsuchioka H, Kawakatsu N, producing a mineralized extracellular matrix, were Kikuchi A, Yamato MJ, Okano T, Ishikawa I. Application of periodontal ligament cell sheet for periodontal regener- grown in the presence of HERS-CM, an acceleration ation: a pilot study in beagle dogs. J Periodontal Res 2005: in the formation of mineral was detected. Analysis of 40: 245–251. the phenotype at the molecular level indicated a 3. Alvarez-Perez MA, Narayanan S, Zeichner-David M, de novo induction of the expression of bone sialo- Rodriguez Carmona B, Arzate H. Molecular cloning, protein and osteocalcin, two markers of mineraliza- expression and immunolocalization of a novel human cementum-derived protein (CP-23). Bone 2006: 38: 409– tion (Fig. 3). These results support the concept that, 419. during root development, the secreted products of 4. Amar S, Chung KM, Nam SH, Karatzas S, Myokai F, Van the Hertwig’s epithelial root sheath induce adjacent Dyke TE. Markers of bone and cementum formation cells of the periodontal ligament to differentiate and accumulate in tissues regenerated in periodontal defects produce new cementum. However, whether these treated with expanded polytetrafluoroethylene mem- branes. J Periodontal Res 1997: 32: 148–158. cells differentiate into cementoblasts or osteoblasts 5. Anitua E. Plasma rich in growth factors: preliminary results awaits further in vivo experiments. of use in the preparation of future sites for implants. Int J Oral Maxillofac Implants 1999: 14: 529–535. 6. Anusaksathien O, Webb SA, Jin QM, Giannobile WV Conclusions Platelet-derived growth factor gene delivery stimulates ex vivo gingival repair. Tissue Eng 2003: 9: 745–756. 7. Anusaksathien O, Jin Q, Zhao M, Somerman MJ, Giannobile It is obvious that major progress has occurred in the WV. Effect of sustained gene delivery of platelet-derived world of biology, medicine and in the past growth factor or its antagonist (PDGF-1308) on tissue- 30 years, and the management of periodontal disease engineered cementum. J Periodontol 2004: 75: 429–440. has benefited from these advances. New knowledge 8. Arweiler NB, Auschill TM, Donos N, Sculean A. Antibac- about the etiology and pathogenesis of periodontitis, terial effect of an enamel matrix protein derivative on in vivo dental biofilm vitality. Clin Oral Invest 2002: 6: 205– the relationship of the disease to systemic problems, 209. and advances in genetics, molecular biology, cell 9. Arzate H, Portilla-Robertson J, Aguilar-Mendoza ME. biology and biomaterials, have opened the door for Recombination of epithelial root sheath and new regenerative techniques based upon the tissue cells in vitro. Arch Med Res 1996: 27: 573–577. engineering approach. Treatment of periodontal 10. Atkinson ME. The development of the mouse molar periodontium. J Periodontal Res 1972: 7: 255–260. disease has evolved from just fighting bacteria to a 11. BarKana I, Narayanan AS, Grosskop A, Savion N, Pitaru S. combined effort to eliminate the offending microor- Cementum attachment protein enriches putative cemen- ganisms, to arrest the progression of tissue damage toblastic populations on root surfaces in vitro. J Dent Res and to regenerate lost tissues. Although some of the 2000: 79: 1482–1488. regenerative techniques have been available for sev- 12. Bartold PM, Narayanan AS, Page RC. Platelet-derived growth factor reduces the inhibitory effects of lipopoly- eral years, and some have shown promising results, saccharide on gingival fibroblast proliferation. J Periodon- none of the techniques are without problems and tal Res 1992: 27: 499–505. none have proven to be 100% effective. Many of the 13. Beertsen W, McCulloch CAG, Sodek J. The periodontal regenerative approaches reviewed in this article are ligament: a unique, multifunctional connective tissues. still under assessment and further research is needed Periodontol 2000 1997: 13: 20–40. to develop cell-based tissue strategies, perhaps using 14. Berkovitz BK. Periodontal ligament: structural and clinical correlates. Dent Update 2004: 31: 46–50. stem cells and biomaterials for delivery of these cells. 15. Birek C, Heersche JN, Jez D, Brunette DM. Secretion of New scaffold materials, which are being developed, a bone resorbing factor by epithelial cells cultured are also needed to address some of the delivery issues from porcine rests of Malassez. J Periodontal Res 1983: 18: (164). What may be concluded from the current sta- 75–81. tus of periodontal regeneration is that, as many 16. Birkedal-Hansen H, Butler WT, Taylor RE. Proteins from the periodontium. characterization of the insoluble of investigators have previously stated, there is not bovine cementum. Calcif Tiss Res 1977: 23: 39–44. going to be one magic solution that can be used to 17. Boabaid F, Gibson CW, Kuehl MA, Berry JE, Snead ML, treat all periodontal patients, but rather a combina- Nociti FH Jr, Katchburian E, Somerman MJ. Leucine-rich tion of different approaches that can be adjusted to amelogenin peptide: a candidate signaling molecule dur- fit the specific need of individual patients. ing cementogenesis. J Periodontol 2004: 75: 1126–1136.

209 Zeichner-David

18. Borrell LN, Papapanou PN. Analytical epidemiology of wounded rat periodontium. J Periodontal Res 2003: 38: periodontitis. J Clin Periodontol 2005: 32: 132–158. 164–174. 19. Bosshardt DD. Are cementoblasts a subpopulation of oste- 36. Cho MI, Lin WL, Genco RJ. Platelet-derived growth factors- oblasts or a unique phenotype? J Dent Res 2005: 84: 390–406. modulated guided tissue regenerative therapy. J Perio- 20. Bosshardt DD, Nanci A. Immunodetection of enamel- and dontol 1995: 66: 522–530. cementum-related (bone) proteins at the enamel-free area 37. Cochran DL, Wozney JM. Biological mediators for perio- and cervical portion of the tooth in rat molars. J Bone dontal regeneration. Periodontol 2000 1999: 19: 40–58. Miner Res 1997: 12: 367–379. 38. D’Errico JA, Macneil RL, Takata T, Berry J, Strayhorn C, 21. Bosshardt DD, Selvig KA. Dental cementum: the dynamic Somerman MJ. Expression of bone associated markers by tissue covering the root. Periodontol 2000 1997: 13: 41– tooth root lining cells, in situ and in vitro. Bone 1997: 20: 117– 75. 126. 22. Bosshardt DD, Sculean A, Windisch P, Pjetursson BE, Lang 39. D’Souza RN, Cavender A, Sood R, Tarnuzzer R, Dickinson NP. Effects of enamel matrix proteins on tissue formation DP, Roberts A, Letterio J. Dental abnormalities in mice along the roots of human teeth. J Periodontal Res 2005: 40: lacking a functional transforming factor-beta 1 (TGF-b1) 158–167. gene indicate a role for TGF-b1 in biomineralization. Int J 23. Boyan LA, Bhargava G, Nishimura F, Orman R, Price R, Oral Biol 1998: 23: 119–131. Terranova VP. Mitogenic and hemotactic responses of 40. Dassule HR, Lewis P, Bei M, Maas R, McMahon AP. Sonic human periodontal ligament cells to the different isoforms hedgehog regulates growth and morphogenesis of the of platelet-derived growth factor. J Dent Res 1994: 73: 1593– tooth. Development 2000: 127: 4775–4785. 1600. 41. Davenport DR, Mailhot JM, Wataha JC, Billman MA, 24. Brice GL, Sampson WJ, Sims MR. An ultrastructural eval- Sharawy MM, Shrout MK. Effects of enamel matrix uation of the relationship between epithelial rests of protein application on the viability, proliferation, and Malassez and orthodontic root resorption and repair in attachment of human periodontal ligament fibroblasts to man. Aust Orthod J 1991: 12: 90–94. diseased root surfaces in vitro. J Clin Periodontol 2003: 25. Bronckers ALJJ, Farach-Carson MC, Van Waveren E, Butler 30: 125–131. WT. Immunolocalization of osteopontin, osteocalcin and 42. Davideau JL, Sahlberg C, Blin C, Papagerakis P, Thesleff I, dentin sialoproteins during dental root formation and early Berdal A. Differential expression of the full-length and cementogenesis in the rat. J Bone Mineral Res 1994: 9: 833– secreted truncated forms of EGF receptor during formation 841. of dental tissues. Int J Dev Biol 1995: 39: 605–615. 26. Browne RM. The pathogenesis of odontogenic cysts: a 43. De Obarrio JJ, Arauz-Dutari JI, Chamberlain TM, Croston review. J Oral Pathol 1975: 4: 31–46. A. The use of autologous growth factors in periodontal 27. Brunette DM. Mechanical stretching increases the number surgical therapy: platelet gel biotechnology – case reports. of epithelial cells synthesizing DNA in culture. J Cell Sci Int J Periodontics Restorative Dent 2000: 20: 486–497. 1984: 69: 35–45. 44. Dennison DK, Vallone DR, Pinero GJ, Rittman B, Caffesse 28. Bunyaratavej P, Wang HL. Collagen membranes: a review. RG. Differential effect of TGF-beta 1 and PDGF on prolif- J Periodontol 2001: 72: 215–229. eration of periodontal ligament cells and gingival fibro- 29. Caffesse RG, Holden MJ, Kon S, Nasjleti CE. The effect of blasts. J Periodontol 1994: 65: 641–648. citric acid and fibronectin application on healing 45. Diekwisch TG. The developmental biology of cementum. following surgical treatment of naturally occurring perio- Int J Dev Biol 2001: 45: 695–706. dontal disease in beagle dogs. J Clin Periodontol 1985: 12: 46. Diekwisch TG. Pathways and fate of migratory cells during 578–590. late tooth organogenesis. Connect Tissue Res 2002: 43: 245– 30. Camelo M, Nevins ML, Schenk RK, Lynch SE, Nevins M. 56. Periodontal regeneration in human Class II furcations 47. Esposito M, Coulthard P, Worthington HV. Enamel matrix using purified recombinant human platelet-derived derivative (Emdogain) for periodontal tissue regeneration growth factor-BB (rhPDGF-BB) with bone allograft. Int J in intrabony defects. Cochrane Database Syst Rev 2003: Periodontics Restorative Dent 2003: 23: 213–225. CD003875. 31. Caton JG, Quinones CR. Etiology of periodontal diseases. 48. Feng F, Hou LT. Treatment of osseous defects with fibro- Curr Opin Dent 1991: 1: 17–28. blast-coated hydroxylapatite particles. J Formos Med Assoc 32. Caton JG, DeFuria EL, Polson AM, Nyman S. Periodontal 1992: 91: 1068–1074. regeneration via selective cell repopulation. J Periodontol 49. Feng F, Liu CM, Hsu WC, Hou LT. Long-term effects of 1987: 58: 546–552. gingival fibroblast-coated hydroxylapatite graft on perio- 33. Cattaneo V, Rota C, Silvestri M, Piacentini C, Forlino A, dontal osseous defects. J Formos Med Assoc 1995: 94: 494– Gallanti A, Rasperini G, Cetta G. Effect of enamel matrix 498. derivative on human periodontal fibroblasts: proliferation, 50. Fetner AE, Hartigan MS, Low SB. Periodontal repair using morphology and root surface colonization. An in vitro PerioGlas in nonhuman primates: clinical and histologic study. J Periodontal Res 2003: 38: 568–574. observations. Compendium. 1994: 15: 935–938. 34. Chai Y, Jiang X, Ito Y, Bringas P, Han J, Rowitch DH, 51. Fong HK, Foster BL, Popowics TE, Somerman MJ. The Soriano P, McMahon AP, Sucov HM. Fate of the mam- crowning achievement: getting to the root of the problem. malian during tooth and mandibular J Dent Educ 2005: 69: 555–570. morphogenesis. Development 2000: 127: 1671–1679. 52. Fowler EB, Breault LG, Cuenin MF. Periodontal disease 35. Chano L, Tenenbaum HC, Lekic PC, Sodek J, McCulloch and its association with systemic disease. Mil Med 2001: CA. Emdogain regulation of cellular differentiation in 166: 85–89.

210 Regeneration of periodontal tissues: cementogenesis revisited

53. Franke Stenport V, Johansson CB. Enamel matrix deriv- 70. Greenstein G. Emdogain: evidence of efficacy. Compend ative and titanium implants. J Clin Periodontol 2003: 30: Contin Educ Dent 2000: 21: 299–305. 359–363. 71. Grzesik WJ, Narayanan AS. Cementum and periodontal 54. Froum SJ, Weinberg MA, Rosenberg E, Tarnow D. A com- wound healing and regeneration. Crit Rev Oral Biol Med parative study utilizing open flap debridement with and 2002: 13: 474–484. without enamel matrix derivative in the treatment of per- 72. Gurpinar A, Onur MA, Cehreli ZC, Tasman F. Effect of iodontal intrabony defects: a 12-month re-entry study. enamel matrix derivative on mouse fibroblasts and marrow J Periodontol 2001: 72: 25–34. stromal osteoblasts. J Biomater Appl 2003: 18: 25–33. 55. Fujiwara N, Tabata MJ, Endoh M, Ishizeki K, Nawa T. 73. Haase HR, Bartold PM. Enamel matrix derivative induces Insulin-like growth factor-I stimulates cell proliferation in matrix synthesis by cultured human periodontal fibroblast the outer layer of Hertwig’s epithelial root sheath and cells. J Periodontol 2001: 72: 341–348. elongation of the tooth root in mouse molars in vitro. Cell 74. Hakki SS, Berry JE, Somerman MJ. The effect of enamel Tissue Res 2005: 320: 69–75. matrix protein derivative on follicle cells in vitro. J Peri- 56. Fujiyama K, Yamashiro T, Fukunaga T, Balam TA, Zheng L, odontol 2001: 72: 679–687. Takano-Yamamoto T. Denervation resulting in dento- 75. Hamamoto Y, Nakajima T, Ozawa H. Ultrastructural and alveolar ankylosis associated with decreased Malassez histochemical study on the morphogenesis of epithelial epithelium. J Dent Res 2004: 83: 625–629. rests of Malassez. Arch Histol Cytol 1989: 52: 61–70. 57. Gao Z, Mackenzie IC, Rittman BR, Korszun AK, Williams 76. Hamamoto Y, Nakajima T, Ozawa H, Uchida T. Production DM, Cruchley AT. Immunocytochemical examination of of amelogenin by enamel epithelium of Hertwig’s root immune cells in periapical granulomata and odontogenic sheath. Oral Surg Oral Med Oral Pathol Oral Radiol Endod cysts. J Oral Pathol 1988: 17: 84–90. 1996: 81: 703–709. 58. Gao J, Symons AL, Bartold PM. Expression of transforming 77. Hamamoto Y, Hamamoto N, Nakajima T, Ozawa H. Mor- growth factor-beta 1 (TGF-beta1) in the developing peri- phological changes of epithelial rests of Malassez in rat odontium of rats. J Dent Res 1998: 77: 1708–1716. molars induced by local administration of N-methy- 59. Gao J, Symons AL, Bartold PM. Expression of transforming lnitrosourea. Arch Oral Biol 1998: 43: 899–906. growth factor-beta receptors types II and III within various 78. Hammarstrom L. Enamel matrix, cementum develop- cells in the rat periodontium. J Periodontal Res 1999: 34: ment and regeneration. J Clin Periodontol 1997: 24: 658– 113–122. 668. 60. Gestrelius S, Lyngstadaas SP, Hammarstrom L. Emdogain– 79. Hammarstrom L, Heijl L, Gestrelius S. Periodontal regen- periodontal regeneration based on biomimicry. Clin Oral eration in a buccal dehiscence model in monkeys after Invest 2000: 4: 120–125. application of enamel matrix proteins. J Clin Periodontol 61. Giannobile WV. Periodontal tissue engineering by growth 1997: 24: 669–677. factors. Bone 1996: 19: 23S–37S. 80. Harrison JW, Roda RS. Intermediate cementum. Develop- 62. Giannobile WV, Somerman MJ. Growth and amelogenin- ment, structure, composition, and potential functions. Oral like factors in periodontal wound healing. A systematic Surg Oral Med Oral Pathol Oral Radiol Endod 1995: 79: 624– review. Ann Periodontol 2003: 8: 193–204. 633. 63. Giannobile WV, Finkelman RD, Lynch SE. Comparison of 81. Hasegawa N, Kawaguchi H, Ogawa T, Uchida T, Kurihara canine and nonhuman primate animal models for perio- H. Immunohistochemical characteristics of epithelial cell dontal therapy. Results following a single administration of rests of Malassez during cementum repair. J Periodontal PDGF/IGF-I. J Periodontol 1994: 65: 1158–1168. Res 2003: 38: 51–56. 64. Giannobile WV, Hernandez RA, Finkelman RD, Ryan S, 82. Hassell TM. Tissues and cells of the periodontium. Kiritsy CP, D’Andrea M, Lynch SE. Comparative effects of Periodontol 2000 1993: 3: 9–38. platelet-derived growth factor-BB and insulin-like growth 83. Heard RH, Mellonig JT. Regenerative materials: an factor-I, individually and in combination, on periodontal overview. Alpha Omegan 2000: 93: 51–58. regeneration in Macaca fascicularis. J Periodontal Res 1996: 84. Heden G, Wennstrom J, Lindhe J. Periodontal tissue 31: 301–12. alterations following Emdogain treatment of periodontal 65. Giannobile WV, Lee CS, Tomala MP, Tejeda KM, Zhu Z. sites with angular bone defects. A series of case reports. Platelet-derived growth factor (PDGF) gene delivery for J Clin Periodontol 1999: 26: 855–860. application in periodontal tissue engineering. J Periodontol 85. Heijl L. Periodontal regeneration with enamel matrix 2001: 72: 815–823. derivative in one human experimental defect. A case 66. Gottlow J. Guided tissue regeneration using bioresorbable report. J Clin Periodontol 1997: 24: 693–696. and non-resorbable devices: initial healing and long-term 86. Helder MN, Karg H, Bervoets TJ, Vukicevic S, Burger EH, results. J Periodontol 1993: 64: 1157–1165. D’Souza RN, Woltgens JH, Karsenty G, Bronckers AL. Bone 67. Gottlow J, Nyman S, Karring T, Lindhe J. New attachment morphogenetic protein-7 (osteogenic protein-1, OP-1) and formation as the result of controlled tissue regeneration. tooth development. J Dent Res 1998: 77: 545–554. J Clin Periodontol 1984: 11: 494–503. 87. Hirooka H. The biologic concept for the use of enamel 68. Graves DT, Kang YM, Kose KN. Growth factors in perio- matrix protein: true periodontal regeneration. Quintessence dontal regeneration. Compend Suppl 1994: 18: S672–S677. Int 1998: 10: 621–630. 69. Graziani F, Laurell L, Tonetti M, Gottlow J, Berglundh T. 88. Ho SP, Sulyanto RM, Marshall SJ, Marshall GW. The Periodontal wound healing following GTR therapy of de- cementum-dentin junction also contains glycosami- hiscence-type defects in the monkey: short-, medium- and noglycans and collagen fibrils. J Struct Biol 2005: 151: long-term healing. J Clin Periodontol 2005: 32: 905–914. 69–78.

211 Zeichner-David

89. Hoang AM, Oates TW, Cochran DL. In vitro wound healing 105. Kagayama M, Sasano Y, Zhu JX, Hirata M, Mizohuchi I, responses to enamel matrix derivative. J Periodontol 2000: Kamakura S. Epithelial rest colocalize with cementoblasts 71: 1270–1277. forming acellular cementum but not with cementoblasts 90. Hoang AM, Klebe RJ, Steffensen B, Ryu OH, Simmer JP, forming cellular cementum. Acta Anat 1998: 163: 1–9. Cochran DL. Amelogenin is a cell adhesion protein. J Dent 106. Kalajzic I, Braut A, Guo D, Jiang X, Kronenberg MS, Mina Res 2002: 81: 497–500. M, Harris MA, Harris SE, Rowe DW. Dentin matrix protein 91. Hou LT, Tsai AY, Liu CM, Feng F. Autologous transplan- 1 expression during osteoblastic differentiation, generation tation of gingival fibroblast-like cells and a hydroxylapatite of an GFP-transgene. Bone 2004: 35: 74–82. complex graft in the treatment of periodontal osseous 107. Kalpidis CD, Ruben MP. Treatment of intrabony perio- defects: cell cultivation and long-term report of cases. Cell dontal defects with enamel matrix derivative: a literature Transplant 2003: 12: 787–797. review. J Periodontol 2002: 73: 1360–1376. 92. Howell TH, Fiorellini JP, Paquette DW, Offenbacher S, 108. Kaneko H, Hashimoto S, Enokiya Y, Ogiuchi H, Shimono Giannobile WV, Lynch SE. A phase I/II clinical trial to M. Cell proliferation and death of Hertwig’s epithelial root evaluate a combination of recombinant human platelet- sheath in the rat. Cell Tissue Res 1999: 298: 95–103. derived growth factor-BB and recombinant human insulin- 109. Karatzas S, Zavras A, Greenspan D, Amar S. Histologic like growth factor-I in patients with periodontal disease. observations of periodontal wound healing after treatment J Periodontol 1997: 68: 1186–1193. with PerioGlas in nonhuman primates. Int J Periodontics 93. Huang KK, Shen C, Chiang CY, Hsieh YD, Fu EJ. Effects of Restorative Dent 1999: 19: 489–499. bone morphogenetic protein-6 on periodontal wound 110. Karlan MS, Hench LL, Madden M, Ogino M. A bone- healing in a fenestration defect of rats. J Periodontal Res bonding bioactive material implant in the head and neck: 2005: 40: 1–10. bioglass. Surg Forum 1978: 29: 575–577. 94. Igarashi R, Sahara T, Shimizu-Ishiura M, Sasaki T. Porcine 111. Kawaguchi H, Hirachi A, Hasegawa N, Iwata T, Hamaguchi enamel matrix derivative enhances the formation of H, Shiba H, Takata T, Kato Y, Kurihara H. Enhancement of reparative dentine and dentine bridges during wound periodontal tissue regeneration by transplantation of bone healing of amputated rat molars. J Electron Microsc 2003: marrow mesenchymal stem cells. J Periodontol 2004: 75: 52: 227–236. 1281–1287. 95. Inoue M, LeGeros RZ, Hoffman C, Diamond K, Rosenberg 112. Kawase T, Okuda K, Yoshie H, Burns DM. Cytostatic action PA, Craig RG. Effect of enamel matrix proteins on the phe- of enamel matrix derivative (EMDOGAIN) on human oral notype expression of periodontal ligament cells cultured on squamous cell carcinoma-derived SCC25 epithelial cells. dental materials. J Biomed Mater Res 2004: 69A: 172–179. J Periodontal Res 2000: 35: 291–300. 96. Ishizaki NT, Matsumoto K, Kimura Y, Wang X, Yamashita 113. Kawase T, Okuda K, Momose M, Kato Y, Yoshie H, Burns A. Histopathological study of dental pulp tissue capped DM. Enamel matrix derivative (EMDOGAIN) rapidly sti- with enamel matrix derivative. J Endod 2003: 29: 176–179. mulates phosphorylation of the MAP kinase family and 97. Ivanovski S, Li H, Haase HR, Bartold PM. Expression of nuclear accumulation of smad2 in both oral epithelial and bone associated macromolecules by gingival and perio- fibroblastic human cells. J Periodontal Res 2001: 36: 367– dontal ligament fibroblasts. J Periodontal Res 2001: 36: 376. 131–141. 114. Kawase T, Okuda K, Yoshie H, Burns DM. Anti-TGF-beta 98. Iwata T, Morotome Y, Tanabe T, Fukae M, Ishikawa I, Oida antibody blocks enamel matrix derivative-induced upreg- S. Noggin blocks osteoinductive activity of porcine enamel ulation of p21WAF1/cip1 and prevents its inhibition of extracts. J Dent Res 2002: 81: 387–391. human oral epithelial cell proliferation. J Periodontal Res 99. Jepsen S, Eberhard J, Herrera D, Needleman I. A systematic 2002: 37: 255–262. review of guided tissue regeneration for periodontal furca- 115. Keila S, Nemcovsky CE, Moses O, Artzi Z, Weinreb M. In tion defects. What is the effect of guided tissue regeneration vitro Effects of Enamel Matrix Proteins on Rat Bone Mar- compared with surgical debridement in the treatment of row Cells and Gingival Fibroblasts. J Dent Res 2004: 83: furcation defects? J Clin Periodontol 2002: 29: 103–116. 134–138. 100. Jernvall J, Thesleff I. Reiterative signaling and patterning 116. King GN. New regenerative technologies: rationale and during mammalian tooth morphogenesis. Mech Dev 2000: potential for periodontal regeneration: 2. Growth factors. 92: 19–29. Dent Update 2001: 28: 60–65. 101. Jiang J, Safavi KE, Spangberg LS, Zhu Q. Enamel matrix 117. King GN. New regenerative technologies: rationale and derivative prolongs primary osteoblast growth. J Endod potential for periodontal regeneration: 1. New advances in 2001: 27: 110–112. established regenerative strategies. Dent Update 2001: 28: 102. Jiang J, Fouad AF, Safavi KE, Spangberg LS, Zhu Q. Effects 7–12. of enamel matrix derivative on gene expression of primary 118. King GN, Cochran DL. Factors that modulate the effects of osteoblasts. Oral Surg Oral Med Oral Pathol Oral Radiol bone morphogenetic protein-induced periodontal regen- Endod 2001: 91: 95–100. eration: a critical review. J Periodontol 2002: 73: 925–936. 103. Jin QM, Anusaksathien O, Webb SA, Rutherford RB, Gian- 119. King GN, Hughes FJ. Bone morphogenetic protein-2 sti- nobile WV. Gene therapy of bone morphogenetic protein mulates cell recruitment and cementogenesis during early for periodontal tissue engineering. J Periodontol 2003: 74: wound healing. J Clin Periodontol 2001: 28: 465–475. 202–213. 120. King GN, King N, Cruchley AT, Wozney JM, Hughes FJ. 104. Jin Q, Anusaksathien O, Webb SA, Printz MA, Giannobile Recombinant human bone morphogenetic protein-2 pro- WV. Engineering of tooth-supporting structures by delivery motes wound healing in rat periodontal fenestration de- of PDGF gene therapy vectors. Mol Ther 2004: 9: 519–526. fects. J Dent Res 1997: 76: 1460–1470.

212 Regeneration of periodontal tissues: cementogenesis revisited

121. King GN, King N, Hughes FJ. Effect of two delivery systems 138. Lynch SE, Ruiz de Castilla G, Williams RC, Kiritsy CP, for recombinant human bone morphogenetic protein-2 on Howell TH, Reddy MS, Antoniades HN. The effects of periodontal regeneration in vivo. J Periodontal Res 1998: short-term application of a combination of platelet-de- 33: 226–236. rived and insulin-like growth factors on periodontal wound 122. Kittel PW, Sampson WJ. RME-induced root resorption and healing. J Periodontol 1991: 62: 458–467. repair: a computerised 3-D reconstruction. Aust Orthod J 139. Lyngstadaas SP, Lundberg E, Ekdahl H, Andersson C, 1994: 13: 144–151. Gestrelius S. Autocrine growth factors in human perio- 123. Kobayashi M, Takiguchi T, Suzuki R, Yamaguchi A, Degu- dontal ligament cells cultured on enamel matrix derivative. chi K, Shionome M, Miyazawa Y, Nishihara T, Nagumo M, J Clin Periodontol 2001: 28: 181–188. Hasegawa K. Recombinant human bone morphogenetic 140. MacNeil RL, Thomas HF. Development of the murine protein-2 stimulates osteoblastic differentiation in cells periodontium. I. role of basement membrane in formation isolated from human periodontal ligament. J Dent Res of a mineralized tissue on the developing root dentin 1999: 78: 1624–1633. surface. J Periodontol 1993: 64: 95–102. 124. Koike H, Uzawa K, Grzesik WJ, Seki N, Endo Y, Kasamatsu 141. MacNeil RL, Thomas HF. Development of the murine A, Yamauchi M, Tanzawa H. GLUT1 is highly expressed in periodontium. II. Role of the epithelial root sheath in for- cementoblasts but not in osteoblasts. Connect Tissue Res mation of the periodontal attachment. J Periodontol 1993: 2005: 46: 117–124. 64: 285–291. 125. Kostopoulos L, Karring T. Susceptibility of GTR-regener- 142. Macneil RL, Berry J, Strayhorn C, Somerman MJ. Expres- ated periodontal attachment to ligature-induced perio- sion of bone sialoprotein mRNA by cells lining the mouse dontitis. J Clin Periodontol 2004: 31: 336–340. tooth root during cementogenesis. Arch Oral Biol 1996: 41: 126. Lambrichts I, Creemers J, Van Steenberghe D. Periodontal 827–835. neural endings intimately relate to epithelial rests of Mal- 143. Madan AK, Kramer B. Immunolocalization of fibroblast assez in humans. A light and electron microscope study. growth factor-2 (FGF-2) in the developing root and sup- J Anat 1993: 182: 153–162. porting structures of the murine tooth. J Mol Histol 2005: 127. Lamster IB. Current concepts and future trends for perio- 36: 171–178. dontal disease and periodontal therapy, Part 2: Classifica- 144. Mariotti A. The extracellular matrix of the periodontium: tion, diagnosis, and nonsurgical and surgical therapy. Dent dynamic and interactive tissues. Periodontol 2000 1993: 3: Today 2001: 20: 86–91. 39–63. 128. Lamster IB, Karabin SD. Periodontal disease activity. Curr 145. Mariotti A Efficacy of chemical root surface modifiers in Opin Dent 1992;2: 39–52. the treatment of periodontal disease. A systematic review. 129. Lang H, Schuler N, Nolden R. Attachment formation fol- Ann Periodontol 2003: 8: 205–226. lowing replantation of cultured cells into periodontal de- 146. Matsuda N, Lin WL, Kumar NM, Cho MI, Genco RJ. fects – a study in minipigs. J Dent Res 1998: 77: 393–405. Mitogenic, chemotactic, and synthetic responses of rat 130. Lekic PC, Rajshankar D, Chen H, Tenenbaum H, McCul- periodontal ligament fibroblastic cells to polypeptide loch CA. Transplantation of labeled periodontal ligament growth factors in vitro. J Periodontol 1992: 63: 515–525. cells promotes regeneration of alveolar bone. Anat Rec 147. Matsuura M, Herr Y, Han KY, Lin WL, Genco RJ, Cho MI. 2001: 262: 193–202. Immunohistochemical expression of extracellular matrix 131. Lekic PC, Nayak BN, Al-Sanea R, Tenenbaum H, Ganss B, components of normal and healing periodontal tissues in McCulloch C. Cell transplantation in wounded mixed the beagle dog. J Periodontol 1995: 66: 579–593. connective tissues. Anat Rec A Discov Mol Cell Evol Biol 148. Maycock J, Wood SR, Brookes SJ, Shore RC, Robinson C, 2005: 287: 1256–1263. Kirkham J. Characterization of a porcine amelogenin pre- 132. Lezot F, Davideau JL, Thomas B, Sharpe P, Forest N, Berdal paration, EMDOGAIN, a biological treatment for perio- A. Epithelial Dlx-2 homeogene expression and cemento- dontal disease. Connect Tissue Res 2002: 43: 472–476. genesis. J Histochem Cytochem 2000: 48: 277–284. 149. McAllister B, Narayanan AS, Miki Y, Page RC. Isolation of a 133. Lindskog S, Hammarstrom L. Formation of intermediate fibroblast attachment protein from cementum. J Perio- cementum III. 3H-tryptophan and 3H-proline uptake into dontal Res 1990: 25: 99–105. the epithelial root sheath of Hertwig in vitro. J Craniofac 150. McCulloch CA Origins and functions of cells essential for Genet Dev Biol 1982: 2: 171–177. periodontal repair: the role of fibroblasts in tissue home- 134. Lindskog S, Blomlof L, Hammarstrom L. Evidence for a role ostasis. Oral Dis 1995: 1: 271–278. of odontogenic epithelium in maintaining the periodontal 151. McCulloch CA, Lekic P, McKee MD. Role of physical forces space. J Clin Periodontol 1988: 15: 371–373. in regulating the form and function of the periodontal 135. Listgarten MA. Similarity of epithelial relationships in the ligament. Periodontol 2000 2000: 24: 56–72. gingiva of rat and man. J Periodontol 1975: 46: 677–680. 152. Melcher AH On the repair potential of periodontal tissues. 136. Lubbock MJ, Harrison VT, Lumsden AG, Palmer RM. J Periodontol 1976: 47: 256–260. Development and cell fate in interspecific (Mus musculus/ 153. Melcher AH. Cells of periodontium: their role in the heal- Mus caroli) intraocular transplants of mouse molar tooth- ing of wounds. Ann R Coll Surg Engl 1985: 67: 130–131. germ tissues detected by in situ hybridization. Arch Oral 154. Mellonig JT Enamel matrix derivative for periodontal re- Biol 1996: 41: 77–84. constructive surgery: technique and clinical and histologic 137. Lukinmaa PL, Mackie EJ, Thesleff I. Immunohistochemical case report. Int J Periodontics Restorative Dent 1999: 19: localization of the matrix glycoprotein-tenascin and the 8–19. ED-sequence form of cellular fibronectin. J Dent Res 1991: 155. Merrilees MJ, Sodek J, Aubin JE. Effects of cells of epithelial 70: 19–26. rests of Malassez and endothelial cells on synthesis of

213 Zeichner-David

glycosaminoglycans by periodontal ligament fibroblasts in 173. Nishimura K, Hayahi M, Matsuda K, Shigeyama Y, vitro. Dev Biol 1983: 97: 146–153. Yamasaki A, Yamaoka A. The chemoattractive potency 156. Metzger Z, Weinstock B, Dotan M, Narayanan AS, Pitaru S. of periodontal ligament, cementum and dentin for Differential chemotactic effect of cementum attachment human gingival fibroblasts. J Periodontal Res 1989: 24: protein on periodontal cells. J Periodontal Res 1998: 33: 146–148. 126–129. 174. Nyman S, Gottlow J, Karring T, Lindhe J. The regenerative 157. Miki Y, Narayanan AS, Page RC. Mitogenic activity of potential of the periodontal ligament. An experimental cementum components to gingival fibroblasts. J Dent Res study in the monkey. J Clin Periodontol 1982: 9: 257–265. 1987: 66: 1399–1403. 175. Oates TW, Rouse CA, Cochran DL. Mitogenic effects of 158. Miletich I, Sharpe PT. Neural crest contribution to mam- growth factors on human periodontal ligament cells in malian tooth formation. Birth Defects Res C Embryo Today vitro. J Periodontol 1993: 64: 142–148. 2004: 72: 200–212. 176. Ohshima M, Nishiyama T, Tokunaga K, Sato S, Maeno M, 159. Miller N, Penaud J, Foliguet B, Membre H, Ambrosini P, Otsuka K. Profiles of cytokine expression in radicular cyst- Plombas M. Resorption rates of 2 commercially available lining epithelium examined by RT-PCR. J Oral Sci 2000: 42: bioresorbable membranes. A histomorphometric study in 239–246. a rabbit model. J Clin Periodontol 1996: 23: 1051–1059. 177. Ohyama M, Suzuki N, Yamaguchi Y, Maeno M, Otsuka K, 160. Mirastschijski U, Konrad D, Lundberg E, Lyngstadaas SP, Ito K. Effect of enamel matrix derivative on the differenti- Jorgensen LN, Agren MS. Effects of a topical enamel matrix ation of C2C12 cells. J Periodontol 2002: 73: 543–550. derivative on skin wound healing. Wound Repair Regen 178. Okubo K, Kobayashi M, Takiguchi T, Takada T, Ohazama 2004: 12: 100–108. A, Okamatsu Y, Hasegawa K. Participation of endogenous 161. Mizutani S, Tsuboi T, Tazoe M, Koshihara Y, Goto S, Togari IGF-I and TGF-beta 1 with enamel matrix derivative-sti- A. Involvement of FGF-2 in the action of Emdogain on mulated cell growth in human periodontal ligament cells. normal human osteoblastic activity. Oral Dis 2003: 9: 210– J Periodontal Res 2003: 38: 1–9. 217. 179. Okuda K, Momose M, Miyazaki A, Murata M, Yokoyama S, 162. Morsczeck C, Gotz W, Schierholz J, Zeilhofer F, Kuhn U, Yonezawa Y, Wolff LF, Yoshie H. Enamel matrix derivative Mohl C, Sippel C, Hoffmann KH. Isolation of precursor in the treatment of human intrabony osseous defects. cells (PCs) from human dental follicle of wisdom teeth. J Periodontol 2000: 71: 1821–1828. Matrix Biol 2005: 24: 155–165. 180. Okuda K, Kawase T, Momose M, Murata M, Saito Y, Suzuki 163. Murphy KG, Gunsolley JC. Guided tissue regeneration for H, Wolff LF, Yoshie H. Platelet-rich plasma contains high the treatment of periodontal intrabony and furcation levels of platelet-derived growth factor and transforming defects. A systematic review. Ann Periodontol 2003: 8: 266– growth factor-beta and modulates the proliferation of 302. periodontally related cells in vitro. J Periodontol 2003: 74: 164. Murphy WL, Mooney DJ. Controlled delivery of inductive 849–857. proteins, plasmid DNA and cells from tissue engineering 181. Orban B. The epithelial network in the periodontal mem- matrices. J Periodontal Res 1999: 34: 413–419. brane. J Am Dent Assoc 1952: 44: 632–635. 165. Nabers CL, Pfeifer JS, Raust GT Jr, Ross SJ. What is the 182. Owens PDA. Ultrastructure of Hertwig’s epithelial root place of bone grafts in periodontal therapy? Periodontal sheath during early root development in premolar teeth in Abstr 1967: 5: 149–153. dogs. Arch Oral Biol 1978: 23: 91–104. 166. Nagano T, Iwata T, Ogata Y, Tanabe T, Gomi K, Fukae M, 183. Owens PDA. A light and electron microscopic study of Arai T, Oida S. Effect of heat treatment on bioactivities of early stages of root surface formation in molar teeth in the enamel matrix derivatives in human periodontal liga- rat. Arch oral Biol 1980: 24: 901–907. ment(HPDL) cells. J Periodontal Res 2004: 39: 249–256. 184. Palioto DB, Coletta RD, Graner E, Joly JC, de Lima AF. The 167. Nakae H, Narayanan S, Raines E, Page RC. Isolation and influence of enamel matrix derivative associated with partial characterization of mitogenic factors from cemen- insulin-like growth factor-I on periodontal ligament fi- tum. Biochemistry 1991: 30: 7047–7052. broblasts. J Periodontol 2004: 75: 498–504. 168. Nakamura Y, Hammarstrom L, Lundberg E, Ekdahl H, 185. Palmer RM, Lubbock MJ. The soft connective tissues of the Matsumoto K, Gestrelius S, Lyngstadaas SP. Enamel matrix gingiva and periodontal ligament: are they unique? Oral derivative promotes reparative processes in the dental Dis 1995: 1: 230–237. pulp. Adv Dent Res 2001: 15: 105–107. 186. Palmer RM, Lumsden AG. Development of periodontal 169. Nakamura Y, Hammarstrom L, Matsumoto K, Lyngstadaas ligament and alveolar bone in homografted recombina- SP. The induction of reparative dentine by enamel pro- tion’s of enamel organs and papillary, pulpal, and follicular teins. Int Endod J 2002: 35: 407–417. mesenchyme in the mouse. Arch Oral Biol 1987: 32: 281– 170. Nakashima M, Reddi AH. The application of bone mor- 289. phogenetic proteins to dental tissue engineering. Nat 187. Parashis A, Tsiklakis K. Clinical and radiographic findings Biotechnol 2003: 21: 1025–1032. following application of enamel matrix derivative in the 171. Nevins M, Camelo M, Nevins ML, Schenk RK, Lynch SE. treatment of intrabony defects. A series of case reports. Periodontal regeneration in humans using recombinant J Clin Periodontol 2000: 27: 705–713. human platelet-derived growth factor-BB (rhPDGF-BB) 188. Park JB, Matsuura M, Han KY, Norderyd O, Lin WL, Genco and allogenic bone. J Periodontol 2003: 74: 1282–1292. RJ. Periodontal regeneration in class III furcation defects in 172. Newman SA, Coscia SA, Jotwani R, Iacono VJ, Cutler CW. beagle dogs using guided tissue regenerative therapy with Effects of enamel matrix derivative on Porphyromonas platelet-derived growth factor. J Periodontol 1995: 66: 462– gingivalis. J Periodontol 2003: 74: 1191–1195. 477.

214 Regeneration of periodontal tissues: cementogenesis revisited

189. Parodi R, Liuzzo G, Patrucco P, Brunel G, Santarelli GA, 206. Ripamonti U, Petit JC, Lemmer J, Austin JC. Regeneration Birardi V, Gaspretto B. Use of Emdogain in the treatment of the connective tissue attachment on surgically exposed of deep intrabony defects: 12-month clinical results. His- roots using fibrin-fibronectin adhesive system. An experi- tologic and radiographic evaluation. Int J Periodontics mental study on the baboon (Papio ursinus). J Periodontal Restorative Dent 2000: 6: 584–595. Res 1987: 22: 320–326. 190. Parodi R, Santarelli GA, Gasparetto B. Treatment of intra- 207. Ripamonti U, Crooks J, Petit JC, Rueger DC. Periodontal bony pockets with Emdogain: results at 36 months. Int J tissue regeneration by combined applications of recom- Periodontics Restorative Dent 2004: 24: 57–63. binant human osteogenic protein-1 and bone morpho- 191. Paynter KJ, Pudy G. A study of the structure, chemical genetic protein-2. A pilot study in Chacma baboons (Papio nature, and development of cementum in the rat. Anat Rec ursinus). Eur J Oral Sci 2001: 109: 241–248. 1958: 131: 233–251. 208. Robey PG, Bianco P, Termine JD. The cellular biology and 192. Peters H, Balling R. Teeth, where and how to make them. molecular biochemistry of bone formation. In: Coe FI, Trends Genet 1999: 15: 59–64. Favuus MJ, editors. Disorders of Bone and Mineral Meta- 193. Phipps RP, Borrello MA, Blieden TMJ. Fibroblast hetero- bolism. New York: Raven Press, 1992: 241–263. geneity in the periodontium and other tissues. J Perio- 209. Rose LF, Rosenberg E. Bone grafts and growth and differ- dontal Res 1997: 32: 159–165. entiation factors for regenerative therapy: a review. Pract 194. Pihlstrom BL, Michalowicz BS, Johnson NW. Periodontal Proced Aesthet Dent 2001: 13: 725–734. diseases. Lancet 2005: 366: 1809–1820. 210. Rutherford RB, Niekrash CE, Kennedy JE, Charette MF. 195. Pitaru S, McCulloch CA, Narayanan AS. Cellular origins Platelet-derived and insulin-like growth factors stimu- and differentiation control mechanisms during periodon- late regeneration of periodontal attachment in monkeys. tal development and wound healing. J Periodontal Res J Periodontal Res 1992: 27: 285–290. 1994: 29: 81–94. 211. Rutherford RB, Ryan ME, Kennedy JE, Tucker MM, 196. Pitaru S, Narayanan SA, Olsen S, Avion N, Hekmati H, Alt I, Charette MF. Platelet-derived growth factor and dexa- Metzger Z. Specific cementum attachment protein methasone combined with collagen matrix induce regen- enhances selectively the attachment and migration of eration of the periodontium in monkeys. J Clin Periodontol periodontal cells to root surfaces. J Periodontal Res 1995: 1993: 20: 537–544. 30: 360–368. 212. Saffar JL, Lasfargues JJ, Cherruau M. Alveolar bone and the 197. Plemons JM, Dill RE, Rees TD, Dyer BJ, Ng MC, Iacopino : the socket that is never stable. Period- AM. PDGF-B producing cells and PDGF-B gene expression ontol 2000 1997: 13: 76–90. in normal gingival and cyclosporine A-induced gingival 213. Sato Y, Kikuchi M, Ohata N, Tamura M, Kuboki Y. En- overgrowth. J Periodontol 1996: 67: 264–270. hanced cementum formation in experimentally induced 198. Plikus MV, Zeichner-David M, Mayer JA, Reyna J, Bringas cementum defects of the root surface with the application P, Thewissen JG, Snead ML, Chai Y, Chuong CM. Mor- of recombinant basic fibroblast growth factor in collagen phoregulation of teeth: modulating the number, size, gel in vivo. J Periodontol 2004: 75: 243–248. shape and differentiation by tuning Bmp activity. Evol Dev 214. Schroeder HE, Listgarten MA. The gingival tissues: the 2005: 7: 440–457. architecture of periodontal protection. Periodontol 2000 199. Price JA, Wright JT, Kula K, Bowden DW, Hart TC. A 1997: 13: 91–120. common DLX3 gene mutation is responsible for tricho- 215. Schwartz Z, Carnes DL Jr, Pulliam R, Lohmann CH, Sylvia dento-osseous syndrome in Virginia and North Carolina VL, Liu Y, Dean DD, Cochran DL, Boyan BD. Porcine fetal families. J Med Genet 1998: 35: 825–828. enamel matrix derivative stimulates proliferation but not 200. Rasperini G, Ricci G, Silvestri M. Surgical technique for differentiation of pre-osteoblastic 2T9 cells, inhibits pro- treatment of infrabony defects with enamel matrix deriv- liferation and stimulates differentiation of osteoblast-like ative (Emdogain): 3 case reports. Int J Periodontics MG63 cells, and increases proliferation and differentiation Restorative Dent 1999: 19: 578–587. of normal human osteoblast NHOst cells. J Periodontol 201. Reichert T, Storkel S, Becker K, Fisher LW. The role of 2000: 71: 1287–1296. osteonectin in development: an immuno- 216. Sculean A, Donos N, Blaes A, Lauermann M, Reich E, Brecx histological study. Calc Tissue Int 1992: 50: 468–472. M. Comparison of enamel matrix proteins and bio- 202. Reynolds MA, Aichelmann-Reidy ME, Branch-Mays GL, absorbable membranes in the treatment of intrabony Gunsolley JC. The efficacy of bone replacement grafts in periodontal defects. A split-mouth study. J Periodontol the treatment of periodontal osseous defects. A systematic 1999: 70: 255–262. review. Ann Periodontol 2003: 8: 227–265. 217. Sculean A, Chiantella GC, Windisch P, Donos N. Clinical 203. Rincon JC, Haase HR, Bartold PM. Effect of Emdogain on and histologic evaluation of human intrabony defects human periodontal fibroblasts in an in vitro wound-heal- treated with an enamel matrix protein derivative (Emdo- ing model. J Periodontal Res 2003: 38: 290–295. gain). Int J Periodontics Restorative Dent 2000: 20: 374–381. 204. Rincon JC, Xiao Y, Young WG, Bartold PM. Enhanced 218. Sculean A, Donos N, Brecx M, Reich E, Karring T. Treat- proliferation, attachment and osteopontin expression by ment of intrabony defects with guided tissue regeneration porcine periodontal cells exposed to Emdogain. Arch Oral and enamel-matrix-proteins. An experimental study in Biol 2005: 50: 1047–1054. monkeys. J Clin Periodontol 2000: 27: 466–472. 205. Ripamonti U, Reddi AH. Tissue engineering, morphogen- 219. Sculean A, Donos N, Brecx M, Karring T, Reich E. Healing of esis, and regeneration of the periodontal tissues by bone fenestration-type defects following treatment with guided morphogenetic proteins. Crit Rev Oral Biol Med 1997: 8: tissue regeneration or enamel matrix proteins. An experi- 154–163. mental study in monkeys. Clin Oral Investig 2000: 4: 50–56.

215 Zeichner-David

220. Sculean A, Auschill TM, Donos N, Brecx M, Arweiler NB. 236. Somerman MJ, Perez-Mera M, Merkhofer RM, Foster RA. Effect of an enamel matrix protein derivative (Emdogain) In vitro evaluation of extracts of mineralized tissues for on ex vivo vitality. J Clin Periodontol 2001: their application in attachment of fibrous tissue. J Period- 28: 1074–1078. ontol 1987: 58: 349–351. 221. Sculean A, Windisch P, Keglevich T, Gera I. Clinical and 237. Spahr A, Lyngstadaas SP, Boeckh C, Andersson C, Podb- histologic evaluation of an enamel matrix protein deriv- ielski A, Haller B. Effect of the enamel matrix derivative ative combined with a bioactive glass for the treatment of Emdogain on the growth of periodontal pathogens in vitro. intrabony periodontal defects in humans. Int J Periodontics J Clin Periodontol 2002: 29: 62–72. Restorative Dent 2005: 25: 139–147. 238. Spouge JD. A new look at the rests of Malassez: a review of 222. Selvig KA. Electron microscopy of Hertwig’s epithelial their embryological origin, anatomy, and possible role in sheath and of early dentin and cementum formation in the periodontal health and disease. J Periodontol 1980: 51: mouse incisor. Acta Odontol Scand 1963: 21: 175–186. 437–444. 223. Sena K, Morotome Y, Baba O, Terashima T, Takano Y, 239. Steele-Perkins G, Butz KG, Lyons GE, Zeichner-David M, Ishikawa I. Gene expression of growth differentiation fac- Kim HJ, Cho MI, Gronostajski RM. Essential role for NFI-C/ tors in the developing periodontium of rat molars. J Dent CTF transcription-replication factor in tooth root devel- Res 2003: 82: 166–171. opment. Mol Cell Biol 2003: 23: 1075–1084. 224. Seo BM, Miura M, Gronthos S, Bartold PM, Batouli S, 240. Sykaras N, Opperman LA. Bone morphogenetic proteins Brahim J, Young M, Robey PG, Wang CY, Shi S. Investiga- (BMPs): how do they function and what can they offer the tion of multipotent postnatal stem cells from human per- clinician? J Oral Sci 2003: 45: 57–73. iodontal ligament. Lancet 2004: 364: 149–155. 241. Talic NF, Evans CA, Daniel JC, Zaki AE. Proliferation of 225. Seo BM, Miura M, Sonoyama W, Coppe C, Stanyon R, Shi epithelial rests of Malassez during experimental tooth S. Recovery of stem cells from cryopreserved periodontal movement. Am J Orthod Dentofacial Orthop 2003: 123: ligament. J Dent Res 2005: 84: 907–912. 527–533. 226. Shear M, Pindborg JJ. Microscopic features of the lateral 242. Tatemoto Y, Okada Y, Mori M. Squamous odontogenic periodontal cyst. Scand J Dent Res 1975: 83: 103–110. tumor: immunohistochemical identification of keratins. 227. Shimizu E, Nakajima Y, Kato N, Nakayama Y, Saito R, Oral Surg Oral Med Oral Pathol 1989: 67: 63–67. Samoto H, Ogata Y. Regulation of rat bone sialoprotein 243. Ten Cate AR. A fine structural study of coronal and root gene transcription by enamel matrix derivative. J Period- in the mouse. Observations of the so called ontol 2004: 75: 260–267. ÔÔvon korff fibersÕÕ and their contribution to mantle dentine. 228. Shimizu E, Saito R, Nakayama Y, Nakajima Y, Kato N, Takai J Anat 1978: 125: 183–197. H, Kim DS, Arai M, Simmer J, Ogata Y. Amelogenin stimu- 244. Ten Cate AR. The role of epithelium in the development, lates bone sialoprotein (BSP) expression through fibroblast structure and function of the tissues of tooth support. Oral growth factor 2 response element and transforming growth Dis 1996: 2: 55–62. factor-beta1 activation element in the promoter of the BSP 245. Ten Cate AR, Mills C. The development of the periodontium: gene. J Periodontol 2005: 76: 1482–1489. the origin of alveolar bone. Anat Rec 1972: 173: 69–78. 229. Shimizu-Ishiura M, Tanaka S, Lee WS, Debari K, Sasaki 246. Tenorio D, Cruchley A, Hughes FJ. Immunocytochemical T. Effects of enamel matrix derivative to titanium investigation of the rat cementoblast phenotype. J Perio- implantation in rat femurs. J Biomed Mater Res 2002: 60: dontal Res 1993: 28: 411–419. 269–276. 247. Terranova VP. Periodontal and bone regeneration factor, 230. Shroff B, Kashner JE, Keyser JD, Hebert C, Norris K Epi- materials and methods. International patent # WO 90/ dermal growth factor and epidermal growth factor-recep- 100017. 1990. tor expression in the mouse dental follicle during tooth 248. Terranova VP, Wikesjo UM. Extracellular matrices and eruption. Arch Oral Biol 1996: 41: 613–617. polypeptide growth factors as mediators of functions of 231. Silvestri M, Rasperini G, Euwe E. Enamel matrix derivative cells of the periodontium. J Periodontol 1987: 58: 371–380. in treatment of infrabony defects. Pract Periodontics 249. Thesleff I. Developmental biology and building a tooth. Aesthet Dent 1999: 11: 615–616. Quintessence Int 2003: 34: 613–620. 232. Slavkin HC. Towards a cellular and molecular under- 250. Thomadakis G, Ramoshebi LN, Crooks J, Rueger DC, standing of periodontics: cementogenesis revisited. J Pe- Ripamonti U. Immunolocalization of Bone Morphogenetic riodontol 1976: 47: 249–255. Protein-2 and -3 and Osteogenic Protein-1 during murine 233. Slavkin HC, Boyd A. Cementum: an epithelial secretory tooth root morphogenesis and in other craniofacial struc- product? J Dent Res 1974: 53: 157. tures. Eur J Oral Sci 1999: 107: 368–377. 234. Slavkin HC, Bringas P, Bessem C, Santos V, Nakamura M, 251. Thomas HF. Root formation. Int J Dev Biol 1995: 39: 231–237. Hsu M, Snead ML, Zeichner-David M, Fincham AG. Her- 252. Thomas HF, Kollar EJ. Differentiation of odontoblasts in twig’s epithelial root sheath differentiation and initial ce- grafted recombinants of murine epithelial root sheath and mentum and bone formation during long-term organ dental mesenchyme. Arch Oral Biol 1989: 34: 27–35. culture of mouse mandibular first molars using serumless, 253. Thomas BL, Tucker AS, Qui M, Ferguson CA, Hardcastle Z, chemically-defined medium. J Periodontal Res 1988: 23: Rubenstein JL, Sharpe P. Role of Dlx-1 and Dlx-2 genes in 28–40. patterning of the murine . Development 1997: 235. Slavkin HC, Bessem C, Fincham AG, Bringas P, Santos V, 124: 4811–4818. Snead ML, Zeichner-David M. Human and mouse ce- 254. Tokiyasu Y, Takata T, Saygin E, Somerman M. Enamel mentum proteins immunologically related to enamel factors regulate expression of genes associated with ce- proteins. Biochim Biophys Acta 1989: 991: 12–18. mentoblasts. J Periodontol 2000: 71: 1829–1839.

216 Regeneration of periodontal tissues: cementogenesis revisited

255. Toyosawa S, Okabayashi K, Komori T, Ijuhin N. mRNA T, Abiko Y. Biological characteristics of the junctional expression and protein localization of dentin matrix pro- epithelium. J Electron Microsc 2003: 52: 627–639. tein 1 during dental root formation. Bone. 2004: 34: 124– 268. Yoneda S, Itoh D, Kuroda S, Kondo H, Umezawa A, Ohya K, 133. Ohyama T, Kasugai S. The effects of enamel matrix deriv- 256. Van der Pauw MT, Van den Bos T, Everts V, Beertsen W. ative (EMD) on osteoblastic cells in culture and bone Enamel matrix-derived protein stimulates attachment of regeneration in a rat defect. J Periodontal Res 2003: periodontal ligament fibroblasts and enhances alkaline 38: 333–342. phosphatase activity and transforming growth factor beta1 269. Yonemura K, Narayanan AS, Miki Y, Page RC, Okada H. release of periodontal ligament and gingival fibroblasts. Isolation and partial characterization of a growth factor J Periodontol 2000: 71: 31–43. from cementum. Bone Mineral Res 1992: 18: 187–198. 257. Venezia E, Goldstein M, Boyan BD, Schwartz Z. The use of 270. Young WG. Growth hormone and insulin-like growth fac- enamel matrix derivative in the treatment of periodontal tor-I in odontogenesis. Int J Dev Biol 1995: 39: 263–272. defects: a literature review and meta-analysis. Crit Rev Oral 271. Yuan K, Chen CL, Lin MT. Enamel matrix derivative Biol Med 2004: 15: 382–402. exhibits angiogenic effect in vitro and in a murine model. 258. Viswanathan HL, Berry JE, Foster BL, Gibson CW, Li Y, J Clin Periodontol 2003: 30: 732–738. Kulkarni AB, Snead ML, Somerman MJ. Amelogenin: a 272. Zeichner-David M. Is there more to enamel matrix proteins potential regulator of cementum-associated genes. J Peri- than biomineralization? Matrix Biol 2001: 20: 307–316. odontol 2003: 74: 1423–1431. 273. Zeichner-David M, Su Z, Chen L, Zakartchenko V, Caton J, 259. Wallace JA, Vergona K. Epithelial restsÕ function in re- Bringas P. Effect of recombinant mouse amelogenin and plantation: is splinting necessary in replantation? Oral Surg ameloblastin on periodontal ligament cell adhesion and Oral Med Oral Pathol 1990: 70: 644–649. proliferation. EJOS (In press). 260. Wang HL, Greenwell H, Fiorellini J, Giannobile W, Offen- 274. Zeichner-David M, Diekwisch T, Fincham A, Lau E, Mac- bacher S, Salkin L, Townsend C, Sheridan P, Genco RJ. Dougall M, Moradian-Oldak J, Simmer J, Snead M, Slavkin Research, Science and Therapy Committee. Periodontal HC. Control of cell differentiation. Int J Develop regeneration. J Periodontol 2005: 76: 1601–1622. Biol 1995: 39: 69–92. 261. Wesselink PR, Beertsen W. The prevalence and distribution 275. Zeichner-David M, Oishi K, Su Z, Zakartchenko V, Chen LS, of Malassez in the mouse molar and their possible role in Arzate H, Bringas P Jr. Role of Hertwig’s epithelial root repair and maintenance of the periodontal ligament. Arch sheath cells in tooth root development. Dev Dyn 2003: 228: Oral Biol 1993: 88: 399–403. 651–663. 262. Wikesjo UME, Claffey N, Christersson LA, Franzetti LC, 276. Zernik JH, Nowroozi N, Liu YH, Maxson R. Development, Genco RJ, Terranova VP. Repair of periodontal furcation maturation, and aging of the alveolar bone. New insights. defects in beagle dogs following reconstructive surgery Dent Clin North Am 1997: 41: 1–15. including root surface de-mineralization with tetracycline 277. Zetterstrom O, Andersson C, Eriksson L, Fredriksson A, hydrochloride and topical fibronectin application. J Clin Friskopp J, Heden G, Jansson B, Lundgren T, Nilveus R, Periodontol 1988: 15: 73–80. Olsson A, Renvert S, Salonen L, Sjostrom L, Winell A, 263. Wu D-Y, Ikezawa K, Parker T, Saito M, Narayanan A-S. Ostgren A, Gestrelius S. Clinical safety of enamel matrix Characterization of a collagenous cementum-derived derivative (EMDOGAIN) in the treatment of periodontal attachment. J Bone Mineral Res 1996: 11: 686–692. defects. J Clin Periodontol 1997: 24: 697–704. 264. Yamamoto H, Cho SW, Kim EJ, Kim JY, Fujiwara N, Jung 278. Zhao M, Xiao G, Berry JE, Franceschi RT, Reddi A, HS. Developmental properties of the Hertwig’s epithelial Somerman MJ. Bone morphogenetic protein 2 induces root sheath in mice. J Dent Res 2004: 83: 688–692. dental follicle cells to differentiate toward a cementoblast/ 265. Yamamoto T, Domon T, Takahashi S, Arambawatta AK, osteoblast phenotype. J Bone Miner Res 2002: 17: 1441– Wakita M. Immunolocation of proteoglycans and bone- 1451. related noncollagenous glycoproteins in developing acel- 279. Zhao M, Berry JE, Somerman MJ. Bone morphogenetic lular cementum of rat molars. Cell Tissue Res 2004: 317: protein-2 inhibits differentiation and mineralization of 299–312. cementoblasts in vitro. J Dent Res 2003: 82: 23–27. 266. Yamashiro T, Tummers M, Thesleff I. Expression of bone 280. Zhao M, Jin Q, Berry JE, Nociti FH Jr, Giannobile WV, morphogenetic proteins and Msx genes during root for- Somerman MJ. Cementoblast delivery for periodontal tis- mation. J Dent Res 2003: 82: 172–176. sue engineering. J Periodontol 2004: 75: 154–161. 267. Yamaza T, Kido MA, Tanaka T, Hashimoto S, Shimono M, Ishikawa T, Enokiya Y, Muramatsu T, Matsuzaka K, Inoue

217