23
Cementum proteins: role in cementogenesis, biomineralization, periodontium formation and regeneration H IGINIO A RZATE ,M ARGARITA Z EICHNER -D AVID &G ABRIELA M ERCADO -C ELIS Destruction of periodontal tissues is a cardinal sign of periodontal disease, and many factors, such as infec- tions, trauma, orthodontic tooth movement and sys- temic and genetic diseases, can contribute to this process. In periodontal disease, bacteria on the sur- face of the teeth produce a chronic inammation of the gingiva. Cells on the surface of the tooth root and the cementum covering the root are destroyed, and the epithelium from the oral mucosa grows down- wards, producing a gingival crevice. Bacteria deposit in this crevice and the ensuing inammatory process may eventually result in the breakdown of periodon- tal tissues, including the cementum, periodontal liga- ment and alveolar bone. The process of periodontal tissue regeneration is initiated at the moment that the damage takes place by the production of growth factors and cytokines by the damaged and inammatory cells. Periodontal treatment can enhance periodontal healing (60). Root planing or root conditioning is being used as a strat- egy to increase mesenchymal cell migration and attachment to the exposed root surface. Treatment with acid, in particular citric acid, has been found to widen the orices of dentinal tubules, thereby accel- erating cementogenesis and enhancing cementum apposition and connective tissue attachment (125). However, when periodontal ligament cells are removed from the cementum or are unable to regen- erate, bone tissue may invade the periodontal liga- ment space and establish a direct connection between the tooth and the wall of the alveolar socket, resulting in ankylosis. This nonexible type of tooth support can lead to loss of function and eventually to resorption of the root (15). Strategies (such as guided tissue regeneration) have been developed to guide and control regeneration using bioresorbable mem- branes (3, 138, 142) and bone grafts (175). Although effective to a certain point, these strategies have the problem that they are not predictable and do not completely restore the architecture of the original periodontium. To achieve complete repair and regen- eration it is necessary to recapitulate the develop- mental process with complete formation of cementum, bone and periodontal ligament bers. The past 20 years of research have seen tremen- dous advances in our knowledge of the cellular and molecular events involved in the process of devel- oping the periodontium. This knowledge has trans- lated into new therapeutic strategies for periodontal regeneration using molecular approaches (45, 67, 110112, 116, 129, 146, 158, 210, 223, 240). Amongst these strategies are the use of growth factors, such as platelet-derived growth factor and insulin-like growth factors (32, 54, 90, 120, 163, 181, 222), trans- forming growth factor-beta1 (127), basic broblast growth factor (191), dexamethasone (181) and bone morphogenetic proteins (109, 114, 154, 176178). It is believed that these molecules are produced dur- ing cementum formation and are then stored in the cementum matrix to induce periodontal ligament regeneration when needed (200). However, one of the problems of application of these factors for peri- odontal repair is the nonspecic activity of some of the factors on different cell lineages and the rapid loss of the topically applied factors over time (15, 120). As our understanding of the structure, function and composition of cementum increases, so does the 211 Periodontology 2000, Vol. 67, 2015, 211–233 © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd Printed in Singapore. All rights reserved PERIODONTOLOGY 2000

Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

  • Upload
    others

  • View
    19

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

Cementum proteins: role incementogenesis, biomineralization,periodontium formation andregenerationHIGINIO ARZATE, MARGARITA ZEICHNER-DAVID & GABRIELA MERCADO-CELIS

Destruction of periodontal tissues is a cardinal sign ofperiodontal disease, and many factors, such as infec-tions, trauma, orthodontic tooth movement and sys-temic and genetic diseases, can contribute to thisprocess. In periodontal disease, bacteria on the sur-face of the teeth produce a chronic inflammation ofthe gingiva. Cells on the surface of the tooth root andthe cementum covering the root are destroyed, andthe epithelium from the oral mucosa grows down-wards, producing a gingival crevice. Bacteria depositin this crevice and the ensuing inflammatory processmay eventually result in the breakdown of periodon-tal tissues, including the cementum, periodontal liga-ment and alveolar bone.

The process of periodontal tissue regeneration isinitiated at the moment that the damage takes placeby the production of growth factors and cytokines bythe damaged and inflammatory cells. Periodontaltreatment can enhance periodontal healing (60). Rootplaning or root conditioning is being used as a strat-egy to increase mesenchymal cell migration andattachment to the exposed root surface. Treatmentwith acid, in particular citric acid, has been found towiden the orifices of dentinal tubules, thereby accel-erating cementogenesis and enhancing cementumapposition and connective tissue attachment (125).However, when periodontal ligament cells areremoved from the cementum or are unable to regen-erate, bone tissue may invade the periodontal liga-ment space and establish a direct connectionbetween the tooth and the wall of the alveolar socket,resulting in ankylosis. This nonflexible type of toothsupport can lead to loss of function and eventually toresorption of the root (15). Strategies (such as guided

tissue regeneration) have been developed to guideand control regeneration using bioresorbable mem-branes (3, 138, 142) and bone grafts (175). Althougheffective to a certain point, these strategies have theproblem that they are not predictable and do notcompletely restore the architecture of the originalperiodontium. To achieve complete repair and regen-eration it is necessary to recapitulate the develop-mental process with complete formation ofcementum, bone and periodontal ligament fibers.

The past 20 years of research have seen tremen-dous advances in our knowledge of the cellular andmolecular events involved in the process of devel-oping the periodontium. This knowledge has trans-lated into new therapeutic strategies for periodontalregeneration using molecular approaches (45, 67,110–112, 116, 129, 146, 158, 210, 223, 240). Amongstthese strategies are the use of growth factors, suchas platelet-derived growth factor and insulin-likegrowth factors (32, 54, 90, 120, 163, 181, 222), trans-forming growth factor-beta1 (127), basic fibroblastgrowth factor (191), dexamethasone (181) and bonemorphogenetic proteins (109, 114, 154, 176–178). Itis believed that these molecules are produced dur-ing cementum formation and are then stored in thecementum matrix to induce periodontal ligamentregeneration when needed (200). However, one ofthe problems of application of these factors for peri-odontal repair is the nonspecific activity of some ofthe factors on different cell lineages and the rapidloss of the topically applied factors over time (15,120).

As our understanding of the structure, function andcomposition of cementum increases, so does the

211

Periodontology 2000, Vol. 67, 2015, 211–233 © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Printed in Singapore. All rights reserved PERIODONTOLOGY 2000

Page 2: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

potential for new therapies for periodontal regenera-tion using molecules formed by these tissues. Onesuch example has been the use of enamel proteins toinduce cementum and bone regeneration (50, 51, 72–74, 84, 85). A preparation of porcine enamel proteinhas been marketed (Emdogain�) and is currentlybeing used by practicing dentists (42, 52, 55, 105, 237,239). Outcomes from studies using Emdogain suggestthat its clinical action is the result of contaminationwith growth factors such as transforming growth fac-tor-beta (107, 143) and bone morphogenetic proteins(100). However, studies using recombinant amelo-genin, including results obtained in our laboratory,indicate that amelogenin and ameloblastin also havesignaling properties that induce phenotypic changesin cells, and these changes can fluctuate dependingon the target cell (20, 219, 237, 239, 240).

It is the purpose of this review to focus on the roleof cementum and its specific components in the for-mation, repair and regeneration of the periodontium.As cementum is a matrix rich in growth factors thatcould influence the activities of various types of peri-odontal cells, this review will examine the characteris-tics of cementum, its composition and the role ofcementum components, especially the cementumprotein-1, during the process of cementogenesis, andtheir potential usefulness for regeneration of theperiodontal structures in a predictable therapeuticmanner.

What is cementum?

Cementum can be described as the mineralized tis-sue that covers the roots of teeth and serves toattach the tooth to alveolar bone via collagen fibersof the periodontal ligament. Morphological, histo-logical and functional differences appear to existalong the length of the root, leading cementum tobe classified as follows: intermediate cementum(found in the cemento–enamel junction), acellularcementum (found in the coronal and mid-portionsof the root) and cellular cementum (present in theapical and inter-radicular portions of the root con-taining cementoblasts) (21, 33, 71, 75, 183, 187, 207,240).

Studies on dental cementum can be traced back toMalpighi in the 1600s (61). In general, 18th centuryanatomists regarded human teeth as composed onlyof enamel and dentin. However, the studies of Tenonon horses’ teeth, of Blake on elephants’ teeth and ofCuvier on the teeth of many species resulted in therecognition that cementum was a constituent part of

all animal teeth. Initial examination of cementum onthe roots of human teeth is attributed to Ringelmannin 1824, followed by physiologists Jan Evangelista Pur-kinje and his pupils, Fraenkel and Raschkow, in 1835,and later by the classic histologist Anders Adolf Retzi-us, in 1836, who noticed the presence of ‘striaes’ incementum (19, 37, 61, 186). In almost all mammalianspecies the number of incremental structures in thedental cementum (annulations) can be correlatedwith age and are used as such in archeology andforensics (61).

In a very simplistic way we can define cementumas an extracellular matrix composed of calcified col-lagenous Sharpey’s fibrils, collagen, glycosaminogly-cans, proteoglycans and inorganic hydroxyapatite. Inthe same way we can say that the major functionalrole of cementum is to serve as the anatomical struc-tural site for the attachment of Sharpey’s fibers of theperiodontal ligament. However, the more we studythis tissue, the more complex we find its structureand function. Cementum biology goes back to Gott-lieb, in 1942, who stated that “the continuous deposi-tion of cementum layers seems to be of greatimportance and work as a barrier against the down-growth of the epithelium. Newly deposited cemen-tum seems to have the highest vitality and act as thebest barrier. If the ideas about the biology of cemen-tum are correct, it is then our task to find out justhow nature provides for continuous cementum depo-sition and having done so, to imitate the procedure”(62). These studies introduced the notion that notonly does cementum act as a barrier to delimit epi-thelial growth that can impair attachment but alsothat the presence of a continuous cementum layer isnecessary to act as a microbial barrier and thatdefects in this tissue could result in periodontitis (62,63). This idea was further supported by observationsthat the root surfaces of patients with hypophospha-tasia contained areas completely devoid of cemen-tum or covered with a hypoplastic form of acementum-like material. These patients developedearly-onset periodontitis, suggesting that abnormali-ties in the deposition or maintenance of cementumcan result in a defective periodontium highly suscep-tible to microbial invasion and destruction (160).

Based on the different functions attributed tocementum it is clear that a thorough understandingof the biological properties of cementum is requiredto determine its role in periodontal formation andtherefore periodontal regeneration. Furthermore, theprinciples attributed to cementum regenerationmight be used in the regeneration of other mineral-ized tissues.

Arzate et al.

212

Page 3: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

Cementum composition

In order to understand the process of cementogenesisit is important to know the composition of cemen-tum. As in bone and dentin, the major organic com-ponent of cementum is collagen (18). The major typeof collagen is type I collagen, which accounts for 90%of all collagens and plays a structural role during thebiomineralization process, serving as a reservoir forhydroxyapatite nucleation, which successively devel-ops into intrafibrillar apatite crystals (58). Type III col-lagen, which coats type I collagen fibrils, is alsopresent, although in considerably lower quantities. Inaddition to collagens, carboxylated and sulfated mu-copolysaccharides (glycosaminoglycans) are presentin human cementum (217).

Glycosaminoglycans

Glycosaminoglycan-containing proteoglycans areheterogeneous groups of glycoproteins with longrepeating disaccharides. The percentage of glycosa-minoglycans is high in tissues subjected to compres-sive forces, such as cementum. Proteoglycans areknown to interact specifically with collagens in a vari-ety of tissues. It is postulated that proteoglycans incementum are integral components of cell substra-tum attachment matrices and mediate attachmentbetween old and newly formed cementum, thus cre-ating the cementum incremental lines (189, 220).

The major glycosaminoglycans present in humancementum are hyaluronic acid, dermatan sulfate andchondroitin sulfate, and their distribution appears tobe quite different from that reported for soft tissuessuch as gingival connective tissue and periodontal lig-ament, in which dermatan sulfate predominates (12,165). The proteoglycan content of mineralized tissuesis generally relatively low. These differences mightreflect differences in function between hard and softtissue as proteglycans appear to inhibit collagen min-eralization by occupying strategic locations normallydestined to be filled with hydroxyapatite (189). Inbone, dermatan sulfate proteoglycans are orientedparallel to the collagen fiber axis with chondroitin sul-fate proteoglycans and hyaluronic acid, occupying theinterfibrillar region in a space-filling capacity (190).

Amongst other glycosaminoglycans present incementum, keratan sulfate appears to be one of themajor components, which, after digestion with kera-tanase II and endo-beta-galactosidase, produces twocore proteins: lumican and fibromodulin. Interest-ingly, these proteins are localized predominantly innonmineralized cementum (precementum and the

pericementocyte area), suggesting that they playmajor regulatory roles during cementum mineraliza-tion (30). In a similar manner, it was also determinedthat the large chondroitin sulfate glycosaminoglycan,present in cementum, contains the large hyaluronan-binding proteoglycan, versican, and the small intersti-tial proteoglycans, decorin and biglycan. Versican islocalized in lacunae housing cementocytes. Decorinis closely associated with collagen fibers of the peri-odontal ligament and with biglycan in the cemento-blasts/precementum area. The differential tissuedistribution suggests that glycosaminoglycans mayplay distinct roles during the cementogenesis processin addition to regulating the biomineralization ofcementum (31). Syndecan-2 has been found to be sig-nificantly expressed by cells in close contact with theroot surface and within the matrix of reparativecementum, suggesting that it must be associated withcell–matrix interactions during cementummineraliza-tion (224). Biglycan is also associated with the growthof incremental lines in cellular cementum. Further-more, lumican, decorin, versican and biglycan areassociated with the formation of cellular cementumbut not of acellular cementum, suggesting differentcementocyte subpopulations or a differentialresponse of these cells (1). Osteoadherin, a keratin sul-fate-containing proteoglycan, is also associated withthe initial phase of cementum formation because Her-twig’s epithelial root sheath cells express this proteo-glycan during root development (166), and althoughacellular cementum does not contain proteoglycans,initial acellular cementum formation requires a denseaccumulation of proteoglycans (230).

Ostepontin and bone sialoprotein

Cementum contains many noncollagenous proteins,including some major phosphoproteins such as os-teopontin and bone sialoprotein. These proteins playa major role in filling spaces created during collagenassembly and imparting cohesion to the mineral-liketissue by allowing mineral deposition to spread acrossthe entire collagen meshwork (22, 148). The role pro-posed for these proteins is that of regulators ofhydroxyapatite crystal nucleation and growth. It hasbeen suggested that osteopontin and sialoprotein arenecessary for the initiation of crystal formation at thehighly ordered fibrils of type I collagen (179). Both ofthese proteins are acidic: osteopontin has a poly-Aspand sialoprotein contains two poly-Glu domains, therepetitive sequences of which are known to bind cal-cium to mineral surfaces. Osteopontin is presentwithin the periodontal ligament in mature teeth.

Cementum proteins

213

Page 4: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

Sialoprotein and osteopontin remain bound to colla-gen matrix and they possess cell-attachment proper-ties through their arginine-glycine-aspartic acid(RGD) sequences (199). In the periodontium, osteo-pontin is expressed by cells in close contact with acel-lular cementum as well as by cementocytes (26). Ithas also been suggested that osteopontin regulatescell migration, differentiation and survival via interac-tions with avb3 integrin (53). Sialoprotein is also anRGD-containing sialoprotein with cell-attachmentproperties (156) and has a precise spatial associationwith early mineral aggregates, binds strongly tohydroxyapatite and acts as a specific and potentnucleator for hydroxyapatite crystal formation in vitro(95). Cementum contains sialoprotein, which, duringroot formation, is distinctly localized to cells liningthe surface of cementum. It has been suggestedthat sialoprotein modulates the process of ceme-ntogenesis and is involved in the process of chem-oattraction, adhesion and differentiation ofprecementoblasts (121–123, 201). Both sialoproteinand osteopontin are believed to play a role in the dif-ferentiation of cementoblast progenitor cells to ce-mentoblasts (183).

Gla proteins

Matrix gamma-carboxyglutamic acid protein andosteocalcin are the two major Gla-containing pro-teins associated with calcified hard tissues (80–82,172, 173). Both proteins have high affinity for Ca2+

and hydroxyapatite through interaction with the Glaresidue. The distribution of osteocalcin in the mam-malian body is virtually limited to mineralized tis-sues such as bone, dentin and cementum (34). Inthe dental root, osteocalcin expression is localizedin cells lining cellular cementum and acellularcementum. However, cells at the inter-radiculararea also express osteocalcin. In a similar manner,cellular and acellular cementum show expressionof matrix gamma-carboxyglutamic acid protein,although acellular cementum expresses those pro-teins more prominently than does cellular cemen-tum. Matrix gamma-carboxyglutamic acid protein issecreted by cementum-forming cells and is incorpo-rated at the mineralization front (103). One possibleexplanation for the accumulation of matrix gamma-carboxyglutamic acid protein in acellular cementumand the outer surface of cellular cementum couldbe to prevent hypercalcification of the cementumsurface (77, 104). During root development in mice,a high level of osteocalcin mRNA is selectivelyexpressed by cells lining root-surface cementoblasts;

in contrast, osteocalcin mRNA is not expressed inthe periodontal ligament (38).

The role of these proteins has been related to regu-lation of mineralization. Matrix gamma-carboxyglu-tamic acid protein-deficient mice show abnormalmineralization and a lack of acellular cementum(104). It has been suggested that both osteocalcin andmatrix gamma-carboxyglutamic acid protein act asnegative regulators of mineralization because matrixgamma-carboxyglutamic acid protein-deficient micepromote calcification of the aortic walls and valves.Thus, both osteocalcin and matrix gamma-carboxy-glutamic acid protein seem to regulate mineralizationby acting as negative regulators, but to differentextents because osteocalcin also inhibits conversionof brushite to hydroxyapatite (81, 173). Other mole-cules present in the cementum extracellular matrixinclude osteonectin, which, during cementogenesis,is synthesized by cementum-producing fibroblasts,cementoblasts and cementocytes (174). Osteonectin,synthesized by mineralizing cells, can bind hydroxy-apatite and is associated with mineralization (25, 80,96, 126, 135). Based on the observation that low con-centrations of osteonectin delay hydroxyapatite-seeded crystal growth in vitro, it is speculated thatosteonectin also acts as a negative regulator by pre-venting, rather than promoting, matrix mineralization(134, 230).

Alkaline phosphatase

Tissue nonspecific alkaline phosphatase (also knownsimply as alkaline phosphatase) has been studied formore than 80 years and is believed to play an impor-tant role in skeletal mineralization. Alkaline phospha-tase is a membrane-bound glycoprotein enzyme thathydrolyses phosphate groups at alkaline pH and alsoinhibits pyrophosphatase, ATPase and protein phos-phatase activity at neutral pH (94). Alkaline phospha-tase is expressed in most body sites during embryonicdevelopment but is confined to bone, kidney, liverand B-lymphocytes during adult life. The fact that it isexpressed in nonmineralizing tissues suggests that ithas other roles besides those associated with mineral-ization. Amongst some of these other functions, it hasbeen suggested that alkaline phosphatase can regu-late tissue turnover and cell proliferation, differentia-tion and maturation (94, 226). Alkaline phosphatase ishighly expressed in cells of the periodontal ligament(56, 69, 99, 124, 151, 231), where it is thought to playa role in phosphate metabolism and cementum for-mation (13), particularly formation of acellularcementum (14, 65). Tissue nonspecific alkaline

Arzate et al.

214

Page 5: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

phosphatase-deficient mice show defective formationof acellular cementum, which results in very thin andirregular-shaped patches around the bases of theperiodontal ligament fibers. No defects were seen inalveolar bone, periodontal ligament and cellularcementum, suggesting that alkaline phosphatase isessential for the formation of acellular cementum(16). However, observations in humans with hypo-phosphatasia as a result of mutations in the tissuenonspecific alkaline phosphatase gene revealed thatcementum formation is almost completely abolishedfor both acellular and cellular cementum, resulting inpremature tooth loss. This phenotype differs fromthat in tissue nonspecific alkaline phosphatase gene-knockout mice, which showed blockage of acellularcementum formation only (213, 214) and might beexplained by the different types of mutation possiblein the tissue nonspecific alkaline phosphatase geneand the severity of their manifestation in humans.One of the major functions of tissue nonspecific alka-line phosphatase is the hydrolysis of inorganic pyro-phosphate, a potent inhibitor of hydroxyapatiteformation (206). Cementoblasts are specifically sensi-tive to the levels of inorganic pyrophosphate/inor-ganic phosphate within the extracellular matrix (150).Changes in the level of tissue nonspecific alkalinephosphatase protein have a significant effect on thefunction of osteoblasts, and consequently on matrixmineralization, indicating that tissue nonspecificalkaline phosphatase plays key biological roles in themineralization of bone and cementum (89). Onenovel strategy to reduce inorganic pyrophosphateand increase cementum neoformation may center onthe modulation of inorganic pyrophosphate/inor-ganic phosphate in the periodontium, which mayresult in more predictable regeneration of cementum(180).

Cementum-specific proteins

Extracellular matrix from different tissues share manysimilarities and yet have different functional proper-ties that make them unique. These properties couldbe the result of quantitative and/or qualitative differ-ences amongst their components. For years it wasbelieved that different mineralized tissues containspecific molecules not present in any other tissue (i.e.amelogenin in enamel, dentin sialophosphoproteinin dentin, etc.) and which could be considered asmarkers for those tissues. As detection techniquesbecame more sensitive, it was found that many ofthese molecules were also expressed in other tissues,although at considerably lower concentrations, and

therefore could still be considered as specific mark-ers. Several proteins, some of which are considered tobe cementum-specific proteins, have been isolatedfrom cementum and characterized.

Cementum-derived growth factor

It is now well established that mineralized tissues,such as bone and dentin, are excellent reservoirs ofgrowth factors that, when needed, can be released bydemineralization and serve to repair or regenerate tis-sues. In a similar way, it has been shown that extractsof cementum have the ability to promote a range ofbiological activities such as cell migration, adhesion,mitogenic activity and differentiation, which areessential for periodontal regeneration (67). Miki et al.(137) were the first to report the presence of mito-genic activity in cementum obtained from humanteeth. Later, Nakae et al. (144) isolated and character-ized mitogenic factors present in the cementummatrix of bovine teeth. In addition to fibroblastgrowth factor, which binds strongly to heparin,another mitogenic factor with moderate heparinaffinity was present in cementum but not in alveolarbone. This factor was named cementum-derivedgrowth factor and it is the major component incementum, accounting for 70% of the mitogenicactivity extracted from this tissue. The cementum-derived growth factor acts synergistically with epider-mal growth factor and induces many of the signalingpathways associated with mitogenesis (234). Thesepathways include an increased concentration of cyto-solic Ca2+, activation of the protein kinase C cascadeand expression of cellular proto-oncogenes. Addition-ally, cementum-derived growth factor may promotethe migration and growth of progenitor cells, presentin the adjacent structures, toward the dentin matrixand participate in their differentiation into cemento-blasts (130, 170). Further characterization of cemen-tum-derived growth factor revealed that themitogenic activity was associated with a 14-kDa pro-tein that showed some homology to insulin-likegrowth factor-1. Although cementum-derived growthfactor activity was inhibited with insulin-like growthfactor-1 and insulin-like growth factor-1 receptorantibodies, there were some differences between thecanonical insulin-like growth factor-1 and cemen-tum-derived growth factor, thus it was concluded thatcementum-derived growth factor is an insulin-likegrowth factor-1-like molecule (97).

The presence of cementum-derived growth factorand other growth factors in cementum indicates thatcementum has the potential to regulate themetabolism and turnover of surrounding tissues, that

Cementum proteins

215

Page 6: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

cementum could serve as a storage site for thosegrowth-inducing molecules and that the cementumproteins are likely to serve a biological role in promot-ing periodontal regeneration (67, 149).

Cementum attachment protein

In addition to cementum-derived growth factor, ithas been reported that human and bovine cementumcontains potent mediators of cell attachment, andthis biological activity is associated with a 55-kDaprotein species (128, 157). This protein was namedcementum attachment protein and further character-ization by amino-acid sequencing showed the pres-ence of four sequences containing Gly-X-Y repeatstypical of collagen. A 17-amino-acid peptide had 82%homology with a type XII domain, and anotherpeptide had 95% homology with collagen type Ia1.However, cementum attachment protein did notcross-react with antibodies to type I, type V, type XIIand type XIV collagen, and its attachment activitywas lost after treatment with bacterial collagenase.These findings, and the fact that a cementumattachment protein monoclonal antibody localizescementum attachment protein only to cementum (7),suggest that cementum attachment protein mightbe a collagenous-attachment protein localized exclu-sively in cementum (225).

Characterization of a complementary DNA clone forcementum attachment protein isolated from a humancementifying fibroma-derived cell line k-ZAP expres-sion library, revealed a novel alternatively splicedsequence. This sequence encodes a 140-amino-acidprotein that is identical to the first 125 N-terminalamino acids of a truncated isoform of 3-hydroxyacyl-CoA dehydratase-1/protein-tyrosine phosphatase-like(proline instead of catalytic arginine), known asPTPLA (212). The remainder of the C-terminus ofPTPLA/cementum attachment protein is encoded bya read-through of the splice donor site in exon 2, andthe truncation eliminates the PTPLA sequence thathas the signature phosphatase active site motif.Although PTPLA mRNA is widely expressed in manytissues, the PTPLA/cementum attachment proteinmRNA is expressed in cementum cells and only mar-ginally in some periodontal ligament cells in humanteeth. PTPLA/cementum attachment protein was notfound to be expressed in rat teeth (184), suggestingthat there could be some species differences in its dis-tribution. Rat roots are known to overproduce apicalcementum after they reach 8 weeks of age, whereasother species maintain a normal layer of cementum.

Interestingly, the cementum attachment protein isa 55-kDa collagenous protein, whereas the PTPLA/

cementum attachment protein codes for a 15-kDaprotein that has no collagen sequences. This suggeststhat perhaps the cementum attachment protein isstrongly associated with collagen chains in thecementum matrix, possibly through cross-linking,which increases its molecular weight. This possibilityis supported by the observation that immunoprecipi-tates of cementum extracts obtained with severalanti-cementum attachment protein monoclonal anti-bodies contain two protein species migrating at55 kDa and ~ 29 kDa, and one monoclonal antibodycross-reacts with both cementum attachment proteinand type I collagen (A.S. Narayanan, unpublisheddata). Expression of PTPLA/cementum attachmentprotein is limited to cementum and to some cells inthe endosteal spaces of bone. This could be explainedas a result of the presence of precursors of cemento-blasts in the endosteal spaces of alveolar bone (130,133, 198). These cells are thought to traverse throughthe periodontal ligament before reaching and differ-entiating on cementum.

It has been shown that cementum attachment pro-tein binds to hydroxyapatite and more strongly tocementum than to the dentin surface (168). Cemen-tum attachment protein also binds to fibronectin, butbinds 150 times more strongly to hydroxyapatite thanto fibronectin (169). Like the cementum attachmentprotein obtained from cementum, recombinantPTPLA/cementum attachment protein also binds tohydroxyapatite with high affinity (212). These obser-vations suggest that PTPLA/cementum attachmentprotein may play a regulatory role during cementumformation (67, 212). It has also been shown thatcementum attachment protein promotes the attach-ment of gingival fibroblasts, endothelial cells andsmooth muscle cells, but not oral sulcular epithelialcells (157). Bone cells bind more strongly to cemen-tum attachment protein than do periodontal liga-ment cells, which, in turn, bind more strongly tocementum attachment protein compared with gingi-val cells (300% for bone cells, 250% for periodontalligament cells and 150% for gingival cells). Cementumattachment protein-coated root slices promote pref-erential adhesion and differentiation of osteoblasticcells (168, 169, 171). Attachment to cementum attach-ment protein by human gingival fibroblasts is medi-ated primarily by the integrin a5b1 (98). The a5b1integrin has been shown to be involved in variousaspects of development (131), and it is possible thatthe a5b1 integrin may also play an important role incementogenesis and neo-cementogenesis throughinteractions with cementum attachment protein.Cementum attachment protein has the capacity to

Arzate et al.

216

Page 7: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

direct cell migration of alveolar bone cells. Periodon-tal ligament cell populations differ in their capacity torecognize and respond to cementum attachment pro-tein. Fifteen per cent of clones of periodontal liga-ment cell cultures bind strongly to cementumattachment protein and this binding capacity is simi-lar to that of alveolar bone cells, suggesting that theorigin of cementoblastic precursors could be osteo-genic. Therefore, cementum attachment protein maypotentially be used to induce preferential repopula-tion of the root surface by appropriate cells (169).Periodontal ligament cells manifest a higher chemo-tactic response to cementum attachment proteinthan do gingival fibroblasts, and this activity is 24-foldgreater than that of fibronectin (136). Cementumattachment protein binds selectively to periodontalligament cells and supports periodontal ligament cellattachment to root surfaces (171). Selective chemo-taxis and attachment by cementum attachment pro-tein may represent the natural pathway by whichcementoblast progenitors are attracted to the rootsurface during homeostasis and regeneration. Par-tially demineralized cementum does not support epi-thelial outgrowth; however, it enhances migrationtoward and attachment of periodontal connective tis-sue cells to dental surfaces in vitro (168, 169). Thus,exposed collagen fibers might improve the capacity ofperiodontal connective tissue cells to compete withepithelial cells and be the first to attach to and colo-nize the root surface after periodontal surgery (167).If this is the case, biochemical alterations in cemen-tum may explain the loss of soft-tissue attachmentfrom diseased root surfaces and the failure for its suc-cessful restoration. Selective chemotaxis by, andattachment through, cementum attachment proteinmight represent the natural pathway by which ce-mentoblast progenitors are attracted to the root sur-face during homeostasis and regeneration.

It has been reported that the cementum attach-ment protein possesses the capacity to bind peri-odontal ligament progenitor clones and that this isdirectly related to their alkaline phosphatase expres-sion and mineralized-like tissue formation. Thesecharacteristics are found in cementoblastoma-derived cells, which produce mineralized nodules (6,118). A direct correlation between alkaline phospha-tase expression and mineralized-like tissue formationwas detected in clones with high binding capacity tocementum attachment protein (118), indicating thatcementum attachment protein is associated withmineralizing-tissue-forming progenitors in the peri-odontal ligament. The groups of clones that bind tocementum attachment protein and produce

mineralized-like tissue, similar to cellular cementum,represent 7% of the periodontal ligament populationand 15% of the mineralized-tissue forming clonesbelonging to the cementoblastic lineage (118). Thehigh binding capacity of cementum attachment pro-tein, combined with a low constitutive percentage ofalkaline phosphatase expression, is of interestbecause it supports the view that cementoblastoma-derived cells express a low constitutive percentage ofalkaline phosphatase-positive cells and that cemento-blasts express low levels of alkaline phosphatase com-pared with alveolar bone-derived osteoblasts (205).

Adhesion of human gingival fibroblasts to cemen-tum attachment protein stimulates mitogen-activatedprotein kinase activity and induces the expression ofc-fos mRNA; in contrast, protein-tyrosine phosphory-lation and c-fos mRNA were not induced in unat-tached cells. As mitogen-activated protein kinase andc-fos mRNA were not induced in monolayer cultures,it was presumed that these reactions are induced byadhesion and are not necessary for cell adhesion(182). The kinetics of mitogen-activated proteinkinase activation was different for cells attaching tofibronectin or polylysine – c-fos mRNA levelsincreased only half as much in cells attaching tofibronectin and very little in cells attaching to polyly-sine. These data demonstrate that cementum attach-ment protein and other adhesion molecules presentin mineralized tissue matrices induce characteristicsignaling events during adhesion, which may play arole in the recruitment of specific cell types duringwound healing and in mediating their specific biolog-ical functions. This differential response is especiallyimportant in periodontal regeneration, in which epi-thelial cells must be excluded and fibroblasts and ce-mentoblasts are to be selected from a pool of variousprogenitor cells (170). Cementum attachment proteinis likely to play a role in the cell selection process. Themitogen-activated protein kinase kinase/mitogen-activated protein kinase pathway participates incementum attachment protein-mediated fibroblastspreading, but cell attachment and proliferation donot appear to require extracellular signal-regulatedkinase-2. Both cementum attachment protein andfibronectin mediate cell attachment through thesame a5b1 integrin (98); however, there are differ-ences in the signaling mechanisms induced bycementum attachment protein and fibronectin dur-ing cell attachment. There are also differences inattachment and spreading promoted by these sub-strates. These differences may explain why cellsattach, spread and migrate differentially on cemen-tum attachment protein-containing surfaces, and

Cementum proteins

217

Page 8: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

such differences can be expected to play a role in therecruitment of cells needed for the regeneration ofcementum and other periodontal structures duringperiodontal regeneration. Cementum attachmentprotein bound to root surfaces has been shown toenhance the recruitment of putative cementoblasticcells to the root surface in vitro (118). The amount ofcementum attachment protein bound might be anindicator of the commitment of a progenitor clone tothe mineralized-tissue-forming cell lineage. Cemen-tum attachment protein is instrumental in recruitingputative cementoblastic progenitors to the root sur-face and is capable of enhancing their differentiation.Also, cells that migrate and attach to root surfacescoated with cementum attachment protein showhigher expression of alkaline phosphatase, sialopro-tein and cementum attachment protein (118). Thisindicates that cementum attachment protein plays animportant role in promoting the differentiation ofputative cementoblast progenitors. Human fibro-blasts attached onto surfaces containing cementumattachment protein as the only adhesion substratecan synthesize DNA. However, the synthesis requiresthe engagement of integrins, presumably the a5b1 towhich cementum attachment protein binds (98). Thisindicates that in vivo, tooth-root surfaces containingcementum attachment protein as the matrix compo-nent are conducive for cell proliferation. Cell attach-ment to cementum attachment protein inducesimmediate-early G1 phase events, and cyclin D1 levelsincrease in the cells adhered to cementum attach-ment protein alone, even without growth factors. Theexpression of cyclin D1 is regulated by adhesion inthe presence of growth factors, and signal reactionsgenerated by binding cementum attachment proteinto cementum attachment protein receptors induceexpression of cyclin D1 (232). Cementum attachmentprotein affects cell-cycle progression through mecha-nisms that are common to other molecules. Never-theless, differences occur in the type and degree ofinduction of these events (182). Cells differing in thecapacity to bind cementum attachment protein alsodiffer in their ability to form mineralized tissue in cul-ture and to produce cementum attachment protein(11, 118). These observations indicate that substancessuch as cementum attachment protein present in thelocal cementum environment could determine whichcells are recruited and how they differentiate duringnormal homeostasis and wound healing, and whetherthe healing response is repair or regeneration. This isimportant in periodontal regeneration when regener-ation requires new cementum formation and restora-tion of connective tissue attachment (48, 170).

Formation of new cementum with inserted Shar-pey’s fibers on previously exposed root surfaces is anessential process in the regeneration of periodontaltissues. This process requires the selective repopula-tion of exposed root surfaces by cementoblastic andfibroblastic cell lineages that originate within theperiodontal ligament and possibly bone (132). Selec-tive repopulation invokes that the growth, differentia-tion, directed migration and attachment of these celllineages should be specifically regulated in time andspace. These actions could be accomplished bycementum components, such as cementum-derivedgrowth factor, cementum attachment protein andcementum protein-1 (67, 171).

Cementum protein-1

Cementum protein-1 was first isolated from humancementum and human cementoblastoma-derivedconditioned media (8–10). It is expressed from a sin-gle-copy gene as a 26-kDa nascent protein that isextensively modified by post-translational events.The human cementum protein-1 gene contains oneexon, spans 1.4 kb and maps to the short arm of chro-mosome 16 (16p13.3). The primary sequence ofcementum protein-1 was first deduced from a humancomplementary DNA sequence that showed 98%homology with a predicted 247-amino-acid sequencepresent in the Pan troglodytes chromosome 16 (5). Nosimilar sequences have been found so far in otherspecies. Human cementum protein-1 is composed of247 amino acids with a calculated molecular weightof 26 kDa, and it appears to be an alkaline protein(isoelectric point = 9.73), with no signal peptide. Thecementum protein-1 gene product is enriched in pro-line (11.3%), glycine (10.5%), alanine (10.1%), serine(8.9%), leucine (8.1%), threonine and arginine (each7.7%) and contains low levels of tryptophan, asparticacid, isoleucine (each 2.0%) and phenylalanine(1.6%). Tyrosine is not present. The amino-acidsequence indicates that the cementum protein-1 islikely to be a nuclear protein; however, it does nothave DNA-binding motifs. Amino acids 30–110 show48% similarity with the human collagen a I (I) chain,46% similarity with type XI and 40% similarity withtype X.

The full-length recombinant cementum protein-1expressed in human-derived gingival fibroblasts ismainly composed of beta-sheet with 10% alpha-helix,32.4% anti-parallel, 5.8% parallel, 16.7% beta-turnand 35% random coil. This feature is associated withproteins that have a high percentage of random coilstructure, which have been shown to be multifunc-tional and to have diverse binding properties; such

Arzate et al.

218

Page 9: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

proteins include SIBLING and HMGI(Y) (35, 41). Thismight help to explain why cementum protein-1 regu-lates crystal growth and composition of apatite crys-tals (4). According to in silico analysis, cementumprotein-1 possesses two N-glycosylation sites, namelyasparagine-X-serine in amino acids 20 and 25 – andthis is consistent with its shift from a protein size ofMr 50,000 to a protein size of Mr 39,000. The preciserole of attached carbohydrates in cementum protein-1 is unknown; however, glycosylation may affect thefunction of cementum protein-1 during the minerali-zation process because their anionic surface can binda large number of Ca2+ ions and regulate hydroxyapa-tite crystal growth (29). Glycans are also implicated inthe regulation of endochondral ossification, boneremodeling and fracture healing (66). Cementum pro-tein-1 appears to be a phosphorylated proteinbecause antibodies against phosphor-serine andphosphor-threonine cross-react with cementum pro-tein-1. The presence of phosphate also favors thebinding of Ca2+ to the protein (102, 209), and proteins(such as sialoprotein and osteopontin) associatedwith the mineralization process are highly phosphor-ylated at threonine and serine residues (236). Thus,cementum protein-1 may play a role at the earlystages of mineralization during the formation of octa-calcium phosphate (Fig. 1). The cementum protein-1does not react with sulfhydryl groups; therefore, allcysteine residues in cementum protein-1 might belinked to disulfide bridges, which generally play a rolestabilizing protein structure (87, 88, 106).

In a steady-state system with the concentrations ofcalcium and phosphate maintained below the thresh-

old of spontaneous precipitation, recombinanthuman-cementum protein-1 is effective in promotingthe nucleation of octacalcium phosphate in an aga-rose gel. It was calculated that as little as 1.0 lg/mL ofcementum protein-1 can promote nucleation (218).Human recombinant cementum protein-1 possesseshigh affinity for hydroxyapatite, even without post-translational modifications, and it affects the mor-phology of apatite crystals. Human cementum pro-tein-1 induces the formation of polymorphouscrystals, as confirmed by X-ray diffraction. Elementalanalysis performed with energy-dispersive X-rayanalysis identified a calcium/phosphorus ratio of 1.4,which corresponds to octacalcium phosphate (Fig. 2).These findings indicate that biologically activecementum protein-1 plays a role during the biomin-eralization process and is required for the synthesis ofneedle-like shape crystals. Octacalcium phosphate isfound to be a transient phase during the growth ofbiological crystals. In small crystals, octacalciumphosphate is completely transformed into hydroxyap-atite by hydrolysis and can only be detected in largecrystals because of its slow kinetics of transformation.Octacalcium phosphate has also been presumed tobe a necessary precursor of biological apatites.

Further characterization of cementum protein-1using western blots with extracted proteins fromhuman cementoblastoma and antibody to periodon-tal ligament-derived cells showed the presence ofthree components: 55-, 50- and 26-kDa species. Theseproducts might represent differences in the degree ofpost-translational modifications, particularly phos-phorylation and glycosylation (5). In vitro studies

A B

C D

Fig. 1. Scanning electron micro-graph images of crystal growth. (A)Human recombinant cementumprotein-1 (20 lg in a 0.5% agarosegel) induced the formation of asphere with irradiating prismaticcrystals. (B) Enlargement of the boxin panel A shows the octacalciumphosphate prismatic crystals. (C)The internal part of the sphereshows a central nucleus with irradi-ating prismatic crystal. (D) A moredetailed view of the box in panel Cshows that the irradiating crystalsare originating from a centralnucleus in a needle-like shape thatlater acquires the prismatic crystalmorphology that grows only incementum protein-1-containinggels.

Cementum proteins

219

Page 10: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

using immunocytochemistry confirmed the expres-sion of cementum protein-1 by cementoblastoma-derived cells and periodontal ligament cells. Almostall (95%) of the cementoblastoma-derived cell popu-lation was positive for cementum protein-1, whereasonly 6% of periodontal ligament cells stained positivefor cementum protein-1. Cementum protein-1 wasalso expressed in a small population (3%) of osteo-blastic cells in vitro, whereas cementum protein-1was not detected in gingival fibroblasts. These smallperiodontal ligament-positive and osteoblast-positivepopulations could represent cementoblast precur-sors, suggesting that cementoblasts and osteoblastsmight have a common ancestor and that cementumprotein-1 could be a marker for the cementoblasticlineage (10). Immunohistochemistry studies using

human periodontal tissues showed localization ofcementum protein-1 throughout the entire cementumsurface, including the cementoid phase of acellularand cellular cementum, cementocytes and cells nearthe blood vessels in the periodontal ligament (Fig. 3).These cells are considered as cementum progenitorcells (7, 8, 10). Cementum protein-1 is not expressed inany other human tissues, indicating that cementumprotein-1 is a tissue-specific protein, restricted to ce-mentoblasts and its progenitor cells, and that it mighthave a role as a local regulator of cell differentiationand extracellular matrix mineralization (4).

An interesting observation was the fact thatcementum protein-1 cross-reacts with antibodies totype X collagen. Collagen type X is a product of thehypertrophic chondrocytes and facilitates endochon-dral ossification (115, 193), which suggests some rela-tionship among cementum protein-1,chondrogenesis and the mineralization processes.This hypothesis is supported by studies showing thathuman cementum extracts promote attachment ofchondrocytes in a dose–dependent manner. Further-more, mesenchymal bud stem cells grown in thepresence of cementum extracts result in the forma-tion of Alcian blue-positive nodules, which synthe-size sulfated proteoglycans (8). The expression ofproteoglycans rich in chondroitin sulfate is charac-teristic of cartilage (28, 40, 185, 211, 221). Takentogether, these studies suggest that cementum-spe-cific proteins can induce stem cells to express a carti-lage phenotype. Furthermore, cementum functionsnot only as an inducer of cell differentiation but alsoas an inducer of proliferation (8).

The function of cementum protein-1 was furthertested by transfection into nonmineralizing cells,such as human gingival fibroblasts. In contrast tonormal human gingival fibroblasts, human gingival fi-broblasts/cementum protein-1 cells showedincreased proliferation, formation of mineralizednodules, increased alkaline phosphatase-specificactivity and the de novo expression of osteocalcin, os-teopontin, sialoprotein, runt-related transcriptionfactor 2/core-binding factor alpha1 and cementumattachment protein mRNAs and protein. These mole-cules are all associated with bone/cementum forma-tion (27). The studies strongly support the notion thatcementum protein-1 has the ability to change the cellphenotype from nonmineralizing (human gingival fi-broblasts) to mineralizing (osteoblast/cementoblast)by regulating proliferation and gene expression,resulting in the differentiation of these cells and theproduction of a mineralized extracellular matrixresembling cementum.

A

B

Fig. 2. (A) Representative energy-dispersive X-ray micro-analysis spectrum of the crystals formed as a result of theeffect of human recombinant cementum protein-1 in asteady-state agarose system. The spectrum shows promi-nent peaks of calcium (Ca) and phosphorus (P) and theCa/P ratio indicates that the crystals are octacalcium phos-phate. C, carbon; Cl, chlorine; O, oxygen; Si, silicon. (B)Direct visualization of cementum protein-1 nanospheres(39 nm high, 81 nm wide, ovoid morphology), as deter-mined by tapping mode atomic force microscopy in air.

Arzate et al.

220

Page 11: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

In-vitro studies have shown that periodontal liga-ment cells can form alkaline phosphatase-positiveand alkaline phosphatase-negative colonies and thatthe alkaline phosphatase-positive periodontal liga-ment cells also express higher levels of mineraliza-tion-related genes (sialoprotein and osteocalcin) thando the alkaline phosphatase-negative cells. Thesedata suggest that alkaline phosphatase-positive peri-odontal ligament cells include osteoblast and/or ce-mentoblast subsets (141). It has been found thatcementum protein-1 is preferentially expressed inalkaline phosphatase-positive cells and that theexpression of cementum protein-1 is reduced whenperiodontal ligament cells are cultured under osteo-genic conditions (i.e. with either bone morphogeneticprotein-2 or in osteogenic induction medium) (113).Overexpression of cementum protein-1 increases theexpression of cementum attachment protein in peri-odontal ligament cells at both mRNA and protein lev-els. The mechanism by which cementum protein-1regulates the expression of cementum attachmentprotein in periodontal ligament cells is not clear.However, the expression of cementum protein-1decreases when cells are committed to osteoblastic/chondroblastic lineages, which suggests that theexpression of cementum protein-1 is being differen-

tially regulated in osteoblasts and cementoblasts andthat knockdown of cementum protein-1 expressionin periodontal ligament cells only affects sialoproteinexpression in cementoblasts, suggesting that cemen-tum protein-1 is associated with the regulation ofsialoprotein expression in cementoblasts (113). Wehave reported the nuclear staining of cementum pro-tein-1 in cementoblasts, whereas periodontal liga-ment cells exhibit intense staining of cementumprotein-1 only in the cytoplasm, indicating that thesubcellular location of cementum protein-1 couldchange during the cementoblastic differentiation ofperiodontal ligament cells. Overexpression of cemen-tum protein-1 in periodontal ligament cells down-regulates periodontal ligament cell markers such asPLAPI/asporin, and increases cementoblast markerssuch as cementum attachment protein and sialopro-tein. These data indicate that cementum protein-1could select periodontal ligament cells, or progenitorcells present in the periodontal ligament, to differen-tiate toward the cementoblastic phenotype (113).

The ability of cementum protein-1 to induce peri-odontal ligament cells to differentiate toward ce-mentoblast/osteoblast and/or chondrogenic-likephenotypes was also tested using a three-dimensionalcell-culture system. Under those conditions, cemen-

CEM CEM CEM

PL PL PL

α-CAP α-CEMP1 MERGE

BV BV BV

CB CB CB

AB AB AB

PVC PVC PVC

α-CEMP1 MERGEα-AMELCB CB CB

ERM

ERM ERM

CEM CEM CEM

A B C

D E F

Fig. 3. Double immunostaining of human periodontal tis-sues with cementum proteins. (A) Immunostaining showsthat anti-bovine CAP IgG1 (a-CAP) cross-reacts with ce-mentoblasts (CB) paravascular cells (PVC) and cell sub-populations in the periodontal ligament (PL). (B)Cementum protein-1 antibody (a-CEMP1) cross-reactsstrongly with CB. (C) Double immunostaining shows thatanti-CAP IgG1 and anti-CEMP1 serum are expressed by CBand PVC in the PL that could represent cementum progen-

itor cells. (D) The epithelial cell rests of Malassez (ERM)and CB cross-react strongly with anti-amelogenin mono-clonal antibody (a-AMEL). (E) The CEMP1 gene product isexpressed by the ERM and CB. (F) Double immunostainingwith a-AMEL and a-CEMP1 show that these proteins areexpressed by similar structures such as the ERM and CB.AB, alveolar bone; BV, blood vessel; CB, cementoblasts;PVC, paravascular cells.

Cementum proteins

221

Page 12: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

tum protein-1 stimulated periodontal ligament cellsto proliferate in a three-dimensional globular mate-rial with morphological features and staining charac-teristics of cartilage and cementum/bone-like tissues.Cementum protein-1 induced periodontal ligamentcells, cultured in a three-dimensional system, toexpress type II collagen and aggrecan (both markersfor pre-hypertrophic chondrocytes) at mRNA andprotein levels (36, 91, 140). Expression of type X colla-gen, a marker of fully differentiated chondrocytes(117), was also increased by the addition of cemen-tum protein-1 to the three-dimensional periodontalligament cell cultures. The expression of SRY (sexdetermining region Y)-box 9, a transcription factorthat mediates chondrogenic differentiation (2, 17), incells grown in the presence of human recombinant-cementum protein-1 suggests that cementum pro-tein-1 promotes chondrogenic differentiation (91).Cementum protein-1 shows sequence similarity withtype X and XI collagens and is immunologicallyrelated to type X collagen (5). It is conceivable thatcementum protein-1 plays a role during the minerali-zation of hypertrophic cartilage and facilitates endo-chondral ossification. Periodontal ligament cells showa multilineage potential to differentiate toward osteo-genic, chondrogenic and adipogenic phenotypes by asignificant up-regulation of cartilage marker genesand osteoblastic differentiation markers (49, 192). Itseems possible that cementum protein-1 exerts a dif-ferentiation role on the periodontal ligament cellpopulation by selecting multipotent stem cells, whichprovide a unique reservoir, to differentiate into vari-ous cell phenotypes (229), or as an inducer of the het-erogeneous periodontal ligament cell populationhaving a differential effect on cells at various degreesof differentiation. This also explains the expression ofcementum protein-1 in periodontal ligament cell sub-populations representing precursors of cemento-blasts or osteoblasts, or both (5, 10, 64). Additionally,there is a direct correlation between the capacity ofhuman periodontal ligament-derived cells to bind toa cementum protein and produce mineralized-liketissue in vitro (11). It has been shown that cementumprotein-1 increases the activity of alkaline phospha-tase. High levels of alkaline phosphatase are alsoassociated with hypertrophic cartilage and bone for-mation, and alkaline phosphatase is considered to bea marker for chondrocyte differentiation because itsactivity appears to be increased during chondrocytehypertrophy (213). The expression of cartilage-forma-tion markers in these cultures suggests that periodon-tal ligament cells have the potential to differentiateinto chondrocytes and then progress rapidly to matu-

ration and bone formation, as suggested by theexpression of collagen type X and sialoprotein.

Studies on cementum regeneration, using a dogmodel for dental pulp necrosis, demonstrated theability of cementum protein-1 to recruit mesenchy-mal stem cells from the periodontal ligament and topromote the proliferation and mineralization of thesecells. In vivo studies co-localized cementum protein-1 and STRO-1 (a marker of mesenchymal stem cells)positive cells adjacent to the root-surface areas whereneocementum is deposited, indicating that the cellsresponsible for reparative cementum deposition areof mesenchymal origin (164). In vitro, cementum pro-tein-1 promotes the proliferation and the migrationof periodontal ligament cells, with the migration frontcomprising STRO-1-positive cells. These studies sug-gest that cementum protein-1 is a mediator in woundhealing and periodontal regeneration because it stim-ulated the proliferation and migration of periodontalligament cells. Cementum protein-1 promotes themigration of STRO-1-positive cells and provides apossible mechanism for the recruitment of mesen-chymal cells through migration toward the cemen-tum protein-1 signal (164). The role of cementumprotein-1 as a chemoattractant and as a promoter ofmineralization is further supported by the findingsthat mineralization is reduced upon blocking cemen-tum protein-1 function in vitro. In cementoblastoma-derived cells, blocking cementum protein-1 activitydecreases alkaline phosphatase activity and theexpression of sialoprotein and osteopontin, but doesnot alter cell proliferation (4). These studies alsoshowed that extracellular calcium increases theexpression of cementum protein-1 and PTPLA/cementum attachment protein in periodontal liga-ment stem cells via the mitogen-activated proteinkinase signaling pathway. This has been confirmed byblocking extracellular signal-regulated kinase-1 andextracellular signal-regulated kinase-e using smallinterfering RNA, which down-regulates the expressionof PTPLA/cementum attachment protein and cemen-tum protein-1. Furthermore, the use of calcium chan-nel blocker prevented the expression of cementumprotein-1 and PTPLA/cementum attachment protein,demonstrating the role for calcium ions in cemento-genesis (164). To date, molecules responsible forrecruiting mesenchymal cells and inducing their dif-ferentiation into cementoblasts have not been identi-fied. These studies suggest that cementum protein-1could be one of the molecules.

In summary, the data presented above stronglyindicate that cementum protein-1 is a unique proteinthat has multiple properties as inducer of mineraliza-

Arzate et al.

222

Page 13: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

tion, proliferation, differentiation and cell matura-tion. Additionally, cementum protein-1 might serveto regulate the mesenchymal stem-cell pool presentin the periodontal ligament and to induce its differen-tiation into different pathways. These properties openthe possibilities of creating cementum protein-1-based therapies for periodontal regeneration.

Enamel-associated proteins in cementum

Many years ago, Slavkin & Boyde (196), proposed ahypothesis that Hertwig’s epithelial root sheath-derived extracellular matrix proteins might be relatedto tooth-crown-derived enamel proteins and thatthese enamel-related proteins might initiate acellularcementum formation. Several human and mousecementum proteins were found to be immunologi-cally related to amelogenin and enamelin. These pro-teins represented species of 72 and 26 kDa that weresecreted by Hertwig’s epithelial root sheath cells(197). A few years later, it was demonstrated that a-meloblastin, an enamel-associated protein, isexpressed by epithelial cells covering the first thinlayer of unmineralized root mantle dentin, and astrong signal is expressed in cells enclosed in the cel-lular cementum known as the epithelial cell rests ofMalassez and in cells in a more coronal position at

the root surface forming acellular cementum (43, 44)(Fig. 4). However, the presence of enamel proteins,especially amelogenins, during root development hasbeen the subject of controversy. Some investigatorsreported the expression of amelogenins by the apicalcells of Hertwig’s epithelial root sheath, whichsecreted small amounts of amelogenins during earlydifferentiation of root development (101), whereasothers did not detect the expression of amelogenin inthese cells (119), only ameloblastin (238). Neverthe-less, a new therapeutic approach, using enamelmatrix derivative to achieve periodontal regeneration,was born, based on the assumption that enamelmatrix proteins synthesized by cells of the Hertwig’sepithelial root sheath could trigger the differentiationof follicle cells into cementoblasts. Specifically, it hasbeen postulated that amelogenin induces the forma-tion of acellular extrinsic fiber cementum. However,others suggest that the tissue formed by treatmentwith enamel matrix derivative results in the formationof a cellular cementum-like tissue or bone with thecharacteristics of cellular intrinsic fiber cementum(23, 24). The bone-like appearance of this tissue is inline with the chondrogenic/osteogenic activity ofenamel matrix derivative (24, 215). Numerous studieshave suggested that enamel matrix derivative can

HERS

H&EH&E

AB

PL

CB

CEM

E G

E

DF

α-CEMP1 α-CEMP1

C

DF

CB

α-CEMP1

HERS

α-CEMP1 α-CEMP1

CB

PL

AB

IEE

OEE

CBA

D F

E

IEE

OEE

Fig. 4. Expression of cementum protein-1 (CEMP1) duringinitial root formation. (A) Inner enamel epithelium (IEE)and outer enamel epithelium (OEE) close to the initial rootformation show strong cross-reactivity with anti-cemen-tum protein-1 serum (a-CEMP1) as well as with a few cellsin the dental follicle (DF). (B) An enlargement of the areain panel A shows that elongated ameloblasts and flat-likeshape cells of the OEE strongly express the CEMP1 gene

product. (C) Cells, possibly cementoblasts (CB), facing theroot surface express CEMP1 and CEMP1 is strong expres-sion of at the cemento–enamel junction. (D) Hertwig0s epi-thelial root sheath (HERS) cells express CEMP1. (E)Hematoxylin and eosin (H&E) staining for orientation. (F)Cells (CB) facing cementum throughout the length of theroot express CEMP1. (G) H&E staining for orientation. AB,alveolar bone; E, enamel; PL, periodontal ligament.

Cementum proteins

223

Page 14: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

have multiple functions, such as the promotion of cellproliferation, differentiation and up-regulation ofextracellular matrix production (36, 50, 51, 83, 86, 139,159, 188, 195, 233). Also, several reports have providedfurther evidence that enamel matrix proteins may beinvolved in root formation (20). Furthermore, amelo-genin null mice showed defects in cementum and adecreased expression of sialoprotein along the rootsurface (57, 219). This is of particular importancebecause sialoprotein acts as a regulator of the miner-alization process in cementum (47, 59).

Amelogenins are the expression products of X andY chromosomal genes and give rise to multiplespliced variants (235). One of these alternativelyspliced variants is a leucine-rich amelogenin peptide(A-4) which has been demonstrated to increase theexpression of osteopontin, sialoprotein and osteopro-tegerin, and to decrease the expression of osteocalcin,in cementoblasts (208, 215, 216). Amelogenin and a-meloblastin can act as signaling molecules in theperiodontal ligament; they have an effect on attach-ment (237) and proliferation of these cells in vitro.Both proteins have a modulatory role and down-regulate the expression of type I collagen, whilstinducing the de novo expression of osteocalcin.Amelogenin also induced the expression of sialopro-tein in periodontal ligament cells, indicating that thisprotein can induce phenotypic changes in these cells(239). Amelogenin has been suggested to have biolog-ical effects on cells of mesenchymal origin, such asperiodontal ligament and gingival fibroblasts, andenamel matrix derivative enhances the growth ofhuman bone marrow stromal cells (68, 93, 215). Inthe past, amelogenin has been shown to be expressedby odontoblasts, periodontal ligament cells, Hertwig’sepithelial root sheath cells and cementoblasts (23, 44,46, 70, 72, 78, 79, 155, 162). Others have described theexpression of amelogenin in hematopoietic stemcells, macrophages and megakaryocytes, rat brainand myoepithelial cells, suggesting a regulatory rolefor amelogenin in the recruitment and differentiationof monocytic cells from the bone marrow towardbecoming mineralized tissue-resorbing cells (boneand cementum osteoclasts/cementoclasts). Amelo-genin, a major structural protein in mineralizingenamel, is also expressed in brain tissue and cells ofthe hematopoietic system (39). Progressive deteriora-tion of cementum is observed in amelogenin knock-out mice and is characterized by the increasedpresence of osteoclasts (78, 79). On the other hand,RT-PCR and western blotting results have demon-strated that Hertwig’s epithelial root sheath cells invitro do not synthesize amelogenin or enamelin, but

they do synthesize ameloblastin. These studiesshowed that Hertwig’s epithelial root sheath cellschange their morphology and produce Von Kossa-positive nodules coincidental with the expression ofdentin matrix protein-1, sialoprotein and osteocalcin,and high levels of alkaline phosphatase activity.Transmission electron microscopy comparison of themineralized extracellular matrix deposited by Her-twig’s epithelial root sheath cells in vitro with acellu-lar cementum deposited in vivo suggests that thisextracellular matrix might be cementum (238).

Hertwig’s epithelial root sheath cells synthesizecementum attachment protein and cementum pro-tein-1 (Fig. 4), thus supporting the idea that Hertwig’sepithelial root sheath cells are capable of producingcementum along with inducing a high activity of alka-line phosphatase. This finding provides further evi-dence that the extracellular matrix deposited by thesecells is acellular cementum, indicating that alkalinephosphatase is a very important component of acel-lular cementum. Hertwig’s epithelial root sheath cellsexpress osteocalcin in vitro, thus indicating the possi-bility that disruption of the basement membrane iscaused by Hertwig0s epithelial root sheath cells whenthey start depositing the acellular cementum. Thesestudies suggested different cellular origins for acellu-lar (Hertwig’s epithelial root sheath cells) and cellular(mesenchymal cementoblasts) cementum (23, 92).Furthermore, Hertwig’s epithelial root sheath cells/epithelial cell rests of Malassez are a unique popula-tion of epithelial cells in the periodontal ligament andare believed to play a crucial role in cementum repair(203). Hertwig’s epithelial root sheath cells/epithelialcell rests of Malassez could differentiate into ce-mentoblasts through epithelial–mesenchymal trans-formation (202). Recently it was demonstrated that invitro Hertwig’s epithelial root sheath/epithelial cellrests of Malassez contain primitive stem cells thatexpress epithelial stem-cell markers such as octamer-binding transcription factor 4, homeobox proteinNANOG and stage-specific embryonic antigen 4(147). These cells might function in creating the bor-der between the ameloblasts and the proliferativeregion of Hertwig’s epithelial root sheath (145) andmight contribute to the formation of cementum and/or enamel repair (76, 194). More recently it wasshown that the epithelial cell rests of Malassez sharesimilar phenotypic and functional characteristics withmesenchymal stem cells and are capable of develop-ing into osteoblasts, adipocytes, chondrocytes andneuron-like cells in vitro (228), similar to thatdescribed in vitro for periodontal ligament stem cells(192, 204). These studies suggest that the epithelial

Arzate et al.

224

Page 15: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

cell rests of Malassez are epithelial stem cells with theability to differentiate into epithelial or mesenchymalcells and play a critical function in periodontalrepair/regeneration.

Dental follicle cells, before the onset of root forma-tion, do not express cementum attachment protein,cementum protein-1 or sialoprotein, which suggeststhe absence of differentiated cells. However, dentalfollicle cells are positive for cementum attachmentprotein and cementum protein-1 when stimulatedwith enamel matrix derivative or bone morphoge-netic protein-2/-7, and their expression was reducedwhen treated with recombinant human-Noggin, awell-known inhibitor of bone morphogenetic proteinactivity (108). Some dental follicle cells express STRO-1, indicating that populations of the dental folliclehave mesenchymal progenitor features (108). Severalinvestigators suggest that the use of enamel matrixderivative enhances the expression of mineralized tis-sue markers (such as alkaline phosphatase) and nod-ule mineralization in dental follicle cells along withthe expression of bone morphogenetic protein-2 andsialoprotein (108). As enamel matrix derivative alsoinduces expression of cementum attachment proteinand cementum protein-1, it is suggested that enamelmatrix derivative promotes the differentiation of den-

tal follicle cells to a cementoblast phenotype ratherthan to an osteoblast phenotype. Recently it wasreported that cementum attachment protein andcementum protein-1 are stringently regulated duringthe cementogenesis process and root formation, andthat expression of cementum attachment protein isinduced more strongly by runt-related transcriptionfactor 2 than is cementum protein-1 (161). Runt-related transcription factor 2 is an important tran-scription factor for osteogenesis and cementogenesis,and is present in the early proliferative osteoblast/ce-mentoblast cell phenotype, a developmental stage atwhich cell proliferation is still required to obtain asufficient number of committed cells for matrix for-mation. Higher expression of cementum attachmentprotein at this early stage of cementogenesis is there-fore consistent with its function of promoting cellproliferation (227). In contrast, cementum protein-1has a more important role during the mineralizationprocess and its relationship is focused at this earlystage to control the mineralization process during ce-mentogenesis (218). Recently we reported that nor-mal human-derived cementoblasts expresscementum attachment protein, cementum protein-1and amelogenin and that they are localized to the cellnucleus. Human cementoblasts express not only

ERM

ERMCEMCEMCEM AB

PL PL

CB

PLCB

BV

BV

BV

BV

BV

CEMCEM CEM

BV

CBCB

PL PLPL

PVC

CB

α-pCKα-AMBN α-CAP

BV

BV

BV

BV

BA

D FE

C

Fig. 5. (A) Hematoxylin and eosin staining showing theperiodontal ligament (PL), cementum (CEM) and alveolarbone (AB). Blood vessels (BV) along with the putative pro-genitor cells of CEM are located close to the CEM surface.(B) Cementoblasts (CB) are ordered in three to four layersof cells facing the CEM surface. (C) The epithelial cell restsof Malassez (ERM) are located in the vicinity of the CB celllayers and the BV, indicating a possible inter-relationshipregulating CEM metabolism and differentiation of CEM

progenitor cells. (D) Ameloblastin (AMBM), an enamel andcementum-related protein, is shown to be expressed by asingle layer of CB facing the CEM surface and paravascularprogenitor cells (PVC) into the PL. (E) A pan-cytokeratinmonoclonal antibody (a-pCK) cross-reacts with CB andsubpopulations of PL cells, possibly supporting the state-ment that CEM is an epithelial product. (F) Cementumattachment protein (CAP) is expressed by subpopulationsof ERM and the CB facing the CEM surface.

Cementum proteins

225

Page 16: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

amelogenin but also other enamel-associated mole-cules, such as ameloblastin, enamelin and tuftelin(Fig. 5). Furthermore, cementum protein-1 inducesthe de-novo expression of amelogenin in periodontalligament cells grown in culture. Perhaps cementumprotein-1 promotes the osteoblast/cementobast phe-notype in periodontal ligament cells through theexpression of amelogenin (91, 152, 153). These data,taken together, suggest that enamel-associated pro-teins and cementum proteins could act synergisticallyin the regulation of cementoblast differentiation andcementum deposition. These data offer newapproaches to determine how the process of ce-mentogenesis is regulated and point out the role ofthese proteins during periodontal homeostasis andrepair/regeneration of periodontal structures, andcould represent new and better therapeuticapproaches for the treatment of periodontal disease.

Conclusions

Detailed knowledge of the biology of cementum iskey for understanding how the periodontium func-tions, identifying pathological issues and for develop-ing successful therapies for repair and regenerationof damaged periodontal tissue. Of particular impor-tance is the regeneration of cementum, a highlysophisticated and complex system, and the intercon-nection of this unique tissue with the neoformationof other periodontal tissues. In this review we havehighlighted the recent advances in our understand-ing of the so-called ‘cementum proteins’ – PTPLA/cementum attachment protein and cementum pro-tein-1 – and their possible role in selecting periodon-tal stem cells, inducing their differentiation andregulating the biological mineralization process asso-ciated with cementum formation. Also, this reviewpoints out the synergistic role of enamel-associatedproteins and cementum proteins in these processes.Although there have been tremendous advances inour knowledge of the identity and function of‘cementum proteins’ at the cell and molecular levels,there is still a great deal left to discover about theseproteins. We are already in a position to explore newalternatives for regenerative techniques based uponthe principles of tissue-engineering methods. Theseproteins and related peptides with biological activityhold great therapeutic potential for the regenerationof not only periodontal structures but also of othermineralized tissues. Nevertheless, this is only thebeginning of our understanding of how these pro-teins might modulate tissue responses in relation to

the regeneration process. What may be concludedfrom the current status of the ‘cementum proteins’is that they can pave the way to establish effectivetherapeutic alternatives to achieve the regenerationof the periodontal structures, and that the impactthat they could have in the field of periodontologyand skeletal tissues looks very promising.

Acknowledgments

This work was supported by DGAPA-UNAMIN216711, IT200414 and by CONACYT 130950. Theauthors are grateful to Professor A. Sampath Naraya-nan of the University of Washington for critical read-ing of the manuscript.

References

1. Ababneh KT, Hall RC, Embery G. The proteoglycans ofhuman cementum: immunohistochemical localization inhealthy, periodontally involved and ageing teeth. J Peri-odontal Res 1999: 34: 87–96.

2. Akiyama H, Chaboissier MC, Martin JF, Schdl A, deCrombrugghe B. The transcription factor Sox9 has essen-tial roles in successive steps of the chondrocyte differenti-ation pathway and is required for expression of Sox5 andSox6. Genes Dev 2002: 16: 2813–2828.

3. Al-Hamdan K, Eber R, Sarment D, Kowalski C, Wang HL.Guided tissue regeneration-based root coverage:meta-analysis. J Periodontol 2003: 74: 1520–1533.

4. Alvarez M, Pitaru S, Alvarez O, Reyes Gasga J, Arzate H.Anti-cementoblastoma-derived protein antibody partiallyinhibits mineralization on a cementoblastic cell line. JStruct Biol 2003: 143: 1–13.

5. Alvarez M, Narayanan S, Zeichner-David M, Carmona B,Arzate H. Molecular cloning, expression and immunolo-calization of a novel human cementum-derived protein(CP-23). Bone 2006: 38: 409–419.

6. Arzate H, Olson SW, Page RC, Narayanan AS. Isolation ofhuman tumor cells that produce cementum proteins inculture. Bone Miner 1992: 18: 15–30.

7. Arzate H, Olson SW, Page RC, Gown AM, Narayanan AS.Production of a monoclonal antibody to an attachmentprotein derived from human cementum. FASEB J 1992: 6:2990–2995.

8. Arzate H, Chimal J, Hern�andez L, D�ıaz de Le�on L.. Humancementumprotein 1 promotes chondrogenesis andmineraliza-tion inmesenchymal cells. J Periodontal Res 1996:31: 144–148.

9. Arzate H, Alvarez M, Aguilar ME, Alvarez O. Humancementum tumor cells have different features fromhuman osteoblastic cells in vitro. J Periodontal Res 1998:33: 249–258.

10. Arzate H, Jim�enez LF, Alvarez M, Landa A, Bar-Kana I,Pitaru S. Immunolocalization of a human cementoblas-toma-conditioned medium-derived protein. J Dent Res2002: 81: 541–546.

11. Bar-Kana I, Savion N, Narayanan AS, Pitaru S. Cementumattachment protein manifestation is restricted to the min-

Arzate et al.

226

Page 17: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

eralized tissue forming cells of the periodontium. Eur JOral Sci 1998: 106: 357–364.

12. Bartold PM, Miki Y, McAllister B, Narayanan AS, Page RC.Glycosaminoglycans of human cementum. J PeriodontalRes 1988: 23: 13–17.

13. Beertsen W, Van den Bos T, Everts V. The possible role ofalkaline phosphatase in acellular cementum formation. JBiol Buccale 1990: 18: 203–205.

14. Beertsen W, Everts V. Formation of acellular root cemen-tum in relation to dental and non-dental hard tissues inthe rat. J Dent Res 1990: 69: 1669–1673.

15. Beertsen W, McCulloch CA, Sodek J. The periodontal liga-ment: a unique, multifunctional connective tissue. Period-ontol 2000 1997: 13: 20–40.

16. Beertsen W, Vandenbos T, Everts V. Root development inmice lacking functional tissue non-specific alkaline phos-phatase gene: inhibition of acellular cementum formation.J Dent Res 1999: 78: 1221–1229.

17. Bi W, Deng JM, Zhang Z, Behringer RR, de CrombruggheB. Sox9 is required for cartilage formation. Nat Genet 1999:22: 85–89.

18. Birkedal-Hansen H, Butler WT, Taylor RE. Proteins of theperiodontium. Characterization of the insoluble collagensof bovine dental cementum. Calcif Tissue Res 1977: 31: 39–44.

19. Blackwood HJ. Intermediate cementum. Br Dent J 1957:102: 345–350.

20. Boabaid F, Gibson CW, Kuehl MA, Berry JE, Snead ML,Nociti FH Jr, Katchburian E, Somerman MJ. Leu-cine-rich amelogenin peptide: a candidate signalingmolecule during cementogenesis. J Periodontol 2004: 75:1126–1136.

21. Bosshardt DD, Selvig KA. Dental cementum: the dynamictissue covering of the root. Periodontol 2000 1997: 13: 41–75.

22. Bosshardt DD, Zalzal S, McKee MD, Nanci A. Develop-mental appearance and distribution of bone sialoproteinand osteopontin in human and rat cementum. Anat Rec1998: 250: 13–33.

23. Bosshardt DD, Nanci A. Hertwig’s epithelial root sheath,enamel matrix proteins, and initiation of cementogenesisin porcine teeth. J Clin Periodontol 2004: 31: 184–192.

24. Bosshardt DD, Sculean A, Windisch P, Pjetursson BE, LangNP. Effects of enamel matrix proteins on tissue formationalong the roots of human teeth. J Periodontal Res 2005: 40:158–167.

25. Bronckers ALJJ, Gay S, DiMuzio MT, Butler WT. Immunol-ocalization of y-carboxyglutamic acid-containing proteinsin developing rat bones. Col Relat Res 1985: 5: 273–281.

26. Bronckers AL, Farach-Carson MC, Van Waveren E, ButlerWT. Immunolocalization of osteopontin, osteocalcin, anddentin sialoprotein during dental root formation and earlycementogenesis in the rat. J Bone Miner Res 1994: 9: 833–841.

27. Carmona B, Alvarez M, Narayanan AS, Zeichner-David M,Reyes J, Molina J, Garc�ıa AL, Su�arez JL, Chavarr�ıa IG, Villar-real E, Arzate H. Human Cementum Protein 1 inducesexpression of bone and cementum proteins by human gin-gival fibroblasts. Biochem Biophys Res Commun 2007: 358:763–769.

28. Carrington JL, Reddi AH. Temporal changes in theresponse of chick limb bud mesodermal cells to trans-

forming growth factor beta-type 1. Exp Cell Res 1990: 186:368–373.

29. Chen CC, Boskey AL. Mechanisms of proteoglycan inhibi-tion of hydroxyapatite growth. Calcif l Tissue Int 1985: 37:395–400.

30. Cheng H, Caterson B, Neame PJ, Lester GE, Yamauchi M.Differential distribution of lumican and fibromodulin intooth cementum. Connect Tissue Res 1996: 34: 87–96.

31. Cheng H, Caterson B, Yamauchi M. Identification and im-munolocalization of chondroitin sulfate proteoglycans intooth cementum. Connect Tissue Res 1999: 40: 37–47.

32. Cho MI, Lin WL, Genco RJ. Platelet-derived growth fac-tor-modulated guided tissue regenerative therapy. J Peri-odontol 1995: 66: 522–530.

33. Cho MI, Garant PR. Development and general structure ofthe periodontium. Periodontol 2000 2000: 24: 9–27.

34. Cole DE, Hanley DA. Osteocalcin. In: Hall BK, editor. Bonematrix and bone specific products. London, UK: CRC Press,1991: 239–294.

35. Cuisinier FJ, Glaisher RW, Voegel JC, Hutchison JL, Br�esEF, Frank RM. Compositional variations in apatites withrespect to preferential ionic extraction. Ultramicroscopy1991: 36: 297–305.

36. Dean DD, Lohmann CH, Sylvia VL, Cochran DL, Liu Y,Boyan BD, Schwartz Z. Effect of porcine fetal enamelmatrix derivative on chondrocyte proliferation, differen-tiation, and local factor production is dependent oncell maturation state. Cells Tissue Organs 2002: 171:117–127.

37. Denton GB. The discovery of cementum. J Dent Res 1939:18: 213–303.

38. D’Errico JA, MacNeil RL, Takata T, Berry J, Strayhorn C,Somerman MJ. Expression of bone associated markers bytooth root lining cells, in situ and in vitro. Bone 1997: 20:117–126.

39. Deutsch D, Haze-Filderman A, Blumenfeld A, Dafni L, Le-iser Y, Shay B, Gruenbaum-Cohen Y, Rosenfeld E, FermonE, Zimmermann B, Haegewald S, Bernimoulin JP, TaylorAL. Amelogenin, a major structural protein in mineralizingenamel, is also expressed in soft tissues: brain and cells ofthe hematopoietic system. Eur J Oral Sci 2006: 114: 183–189.

40. Downie SA, Newman SA. Morphogenetic differencesbetween fore and hind limb precartilage mesenchyme:relation to mechanisms of skeletal pattern formation. DevBiol 1994: 162: 195–208.

41. Dunker AK, Obradovic Z. The protein trinity-linking func-tion and disorder. Nat Biotechnol 2001: 19: 805–806.

42. Esposito M, Coulthard P, Thomsen P, Worthington HV.Enamel matrix derivative for periodontal tissue regenera-tion in treatment of intrabony defects: a Cochrane system-atic review. J Dent Educ 2004: 68: 834–844.

43. Fong CD, Slaby I, Hammarstr€om L. Amelin: an enamel-re-lated protein, transcribed in the cells of epithelial rootsheath. J Bone Miner Res 1996: 11: 892–898.

44. Fong CD, Hammarstr€om L. Expression of amelin and ame-logenin in epithelial root sheath remnants of fully formedrat molars. Oral Surg Oral Med Oral Pathol Oral Radiol En-dod 2000: 90: 218–223.

45. Froum SJ, Gomez C, Breault MR. Current concepts of peri-odontal regeneration. A review of the literature. N Y StateDent 2002: 68: 14–22.

Cementum proteins

227

Page 18: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

46. Fukae M, Tanabe T, Yamakoshi Y, Yamada M, Ujiie Y, OidaS. Immunoblot detection and expression of enamel pro-teins at the apical portion of the forming root in porcinepermanent incisor tooth germs. J Bone Miner Metab 2001:19: 236–243.

47. Ganss B, Kim RH, Sodek J. Bone sialoprotein. Crit Rev OralBiol Med 1999: 10: 79–98.

48. Garrett S. Periodontal regeneration around natural teeth.Ann Periodontol 1996: 1: 621–666.

49. Gay IC, Chen S, MacDougall M. Isolation and characteriza-tion of multipotent human periodontal ligament stemcells. Orthod Craniofacial Res 2007: 10: 149–160.

50. Gestrelius S, Andersson C, Johansson AC, Persson E, Bro-din A, Rydhag L, Hammarstr€om L. Formulation of enamelmatrix derivative for surface coating, kinetics and cell colo-nization. J Clin Periodontol 1997: 24: 678–684.

51. Gestrelius S, Andersson C, Lidstrom D, Hammarstrom L,Somerman M. In vitro studies on periodontal ligamentcells and enamel matrix derivative. J Clin Periodontol 1997:24: 685–692.

52. Gestrelius S, Lyngstadaas SP, Hammarstr€om L. Emdogain–periodontal regeneration based on biomimicry. Clin OralInvestig 2000: 4: 120–125.

53. Giachelli CM, Steitz S. Osteopontin: a versatile regulator ofinflammation and biomineralization. Matrix Biol 2000: 19:615–622.

54. Giannobile WV, Finkelman RD, Lynch SE. Comparison ofcanine and non-human primate animal models for peri-odontal regenerative therapy: results following a singleadministration of PDGF/IGF-I. J Periodontol 1994: 65:1158–1168.

55. Giannobile WV, Somerman MJ. Growth and amelo-genin-like factors in periodontal wound healing. A system-atic review. Ann Periodontol 2003: 8: 193–204.

56. Gibson WA, Fullmer HM. Histochemistry of the periodon-tal ligament. II. The phosphatases. Periodontics 1967: 5:226–232.

57. Gibson CW, Yuan ZA, Hall B, Longenecker G, Chen E, Thy-agarajan T, Sreenath T, Wright JT, Decker S, Piddington R,Harrison G, Kulkarni AB. Amelogenin-deficient mice dis-play an amelogenesis imperfecta phenotype. J Biol Chem2001: 276: 31871–31875.

58. Glimcher MJ. Mechanism of calcification: role of collagenfibrils and collagen-phosphoprotein complexes in vitroand in vivo. Anat Rec 1989: 224: 139–153.

59. Goldberg HA, Warner KJ, Stillman MJ, Hunter GK. Deter-mination of the hydroxyapatite-nucleating region of bonesialoprotein. Connect Tissue Res 1996: 35: 385–392.

60. Goldstein M, Brayer L, Schwartz Z. A critical evaluation ofmethods for root coverage. Crit Rev Oral Biol Med 1996: 7:87–98.

61. Gordon BC. Archaeological tooth and bone seasonal incre-ments: the need for standardized terms and techniques.Archeaozoologia 1993: 2: 9–16.

62. Gottlieb B. Biology of the cementum. J Periodontol 1942:13: 13–19.

63. Gottlieb B. The new concept of periodontoclasia. J Period-ontol 1946: 17: 7.

64. Gould TR, Melcher AH. Migration and division of progeni-tor cell populations in periodontal ligament after wound-ing. J Periodontal Res 1980: 15: 20–42.

65. Groeneveld MC, Everts V, Beertsen W. Formation ofafibrillar acellular cementum-like layers induced byalkaline phosphatase activity from periodontal ligamentexplants maintained in vitro. J Dent Res 1994: 73:1588–1592.

66. Grzesik WJ, Frazier CR, Shapiro JR, Sponseller PD, RobeyPG, Fedarko NS. Age-related changes in human bone pro-teoglycan structure: impact of osteogenesis imperfecta. JBiol Chem 2002: 277: 43638–43647.

67. Grzesik WJ, Narayanan AS. Cementum and periodontalwound healing and regeneration. Crit Rev Oral Biol Med2002: 13: 474–484.

68. Guida L, Annunziata M, Carinci F, Di Feo A, Passaro I,Oliva A. In vitro biologic response of human bone marrowstromal cells to enamel matrix derivative. J Periodontol2007: 78: 2190–2196.

69. Halackov�a Z, Oudr�an L, Kukletov�a M. Localization of someenzymes in the periodontium of the rat molar. Acta Histo-chem 1980: 67: 173–179.

70. Hamamoto Y, Nakajima T, Ozawa H, Uchida T. Productionof amelogenin by enamel epithelium of Hertwig’s rootsheath. Oral Surg Oral Med Oral Pathol Oral Radiol Endod1996: 81: 703–709.

71. Hammarstr€om L, Alatli I, Fong CD. Origins of cementum.Oral Dis 1996: 2: 63–69.

72. Hammarstr€om L. Enamel matrix, cementum developmentand regeneration. J Clin Periodontol 1997: 24: 658–668.

73. Hammarstr€om L. The role of enamel matrix proteins inthe development of cementum and periodontal tissues.Ciba Found Symp 1997: 205: 246–255.

74. Hammarstrom L, Heijl L, Gestrelius S. Periodontal regen-eration in a buccal dehiscence model in monkeys afterapplication of enamel matrix proteins. J Clin Periodontol1997: 24: 669–677.

75. Harrison JW, Roda RS. Intermediate cementum. Develop-ment, structure, composition, and potential functions.Oral Surg Oral Med Oral Pathol Oral Radiol Endod 1995:79: 624–633.

76. Hasegawa N, Kawaguchi H, Ogawa T, Uchida T, KuriharaH. Immunohistochemical characteristics of epithelial cellrests of Malassez during cementum repair. J PeriodontalRes 2003: 38: 51–56.

77. Hashimoto F, Kobayashi Y, Kobayashi ET, Sakai E, Kobay-ashi K, Shibata M, Kato Y, Sakai H. Expression and locali-zation of MGP in rat tooth cementum. Arch Oral Biol 2001:46: 585–592.

78. Hatakeyama J, Sreenath T, Hatakeyama Y, Thyagarajan T,Shum L, Gibson CW, Wright JT, Kulkarni AB. The receptoractivator of nuclear factor-kappa B ligand-mediated osteo-clastogenic pathway is elevated in amelogenin-null mice. JBiol Chem 2003: 278: 35743–35748.

79. Hatakeyama J, Philp D, Hatakeyama Y, Haruyama N,Shum L, Aragon MA, Yuan Z, Gibson CW, Sreenath T,Kleinman HK, Kulkarni AB. Amelogenin-mediatedregulation of osteoclastogenesis, and periodontal cellproliferation and migration. J Dent Res 2006: 85: 144–149.

80. Hauschka PV, Lian JB, Gallop PM. Direct identification ofthe calcium-binding amino acid, gamma-carboxygluta-mate, in mineralized tissue. Proc Natl Acad Sci USA 1975:72: 3925–3929.

Arzate et al.

228

Page 19: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

81. Hauschka PV, Lian JB, Cole DE, Gundberg CM. Osteocal-cin and matrix Gla protein: vitamin K-dependent proteinsin bone. Physiol Rev 1989: 69: 990–1047.

82. Hauschka PV, Wians FH Jr. Osteocalcin-hydroxyapatiteinteraction in the extracellular organic matrix of bone.Anat Rec 1989: 224: 180–188.

83. He J, Jiang J, Safavi KE, Spangberg LS, Zhu Q. Emdogainpromotes osteoblast proliferation and differentiation andstimulates osteoprotegerin expression. Oral Surg Oral MedOral Pathol Oral Radiol Endod 2004: 97: 239–245.

84. Heijl L. Periodontal regeneration with enamel matrixderivative in one human experimental defect. A casereport. J Clin Periodontol 1997: 24: 693–696.

85. Hirooka H. The biologic concept for the use of enamelmatrix protein: true periodontal regeneration. Quintes-sence Int 1998: 29: 621–630.

86. Hoang AM, Klebe RJ, Steffensen B, Ryu OH, Simmer JP,Cochran DL. Amelogenin is a cell adhesion protein. J DentRes 2002: 81: 497–500.

87. Hoang QQ, Sicheri F, Howard AJ, Yang DSC. Bone recogni-tion mechanism of porcine osteocalcin from crystal struc-ture. Nature 2003: 425: 977–980.

88. Hohenester E, Maurer P, Hohenadl C, Timpl R, JansoniusJN, Engel J. Structure of a novel extracellular Ca(2+)-bindingmodule in BM-40. Nat Struct Biol 1996: 3: 67–73.

89. Hough TA, Polewski M, Johnson K, Cheeseman M, NolanPM, Vizor L, Rastan S, Boyde A, Pritzker K, Hunter AJ,Fisher EM, Terkeltaub R, Brown SD. Novel mouse modelof autosomal semidominant adult hypophosphatasia has asplice site mutation in the tissue nonspecific alkalinephosphatase gene Akp2. J Bone Miner Res 2007: 22: 1397–1407.

90. Howell TH, Fiorellini JP, Paquette DW, Offenbacher S, Gi-annobile WV, Lynch SE. A phase I/II clinical trial to evalu-ate a combination of recombinant human platelet-derivedgrowth factor-BB and recombinant human insulin-likegrowth factor-I in patients with periodontal disease. J Peri-odontol 1997: 68: 1186–1193.

91. Hoz L, Romo E, Zeichner-David M, Sanz M, Nu~nez J,Gait�an L, Mercado G, Arzate H. Cementum protein(CEMP1) induces differentiation by human periodontalligament cells under three-dimensional culture. Cell BiolInt 2012: 36: 129–136.

92. Huang X, Bringas P Jr, Slavkin HC, Chai Y. Fate of Her-twig’s epithelial root sheath during tooth root develop-ment. Dev Biol 2009: 334: 22–30.

93. Huang YC, Tanimoto K, Tanne Y, Kamiya T, Kunimatsu R,Michida M, Yoshioka M, Yoshimi Y, Kato Y, Tanne K.Effects of human full-length amelogenin on the prolifera-tion of human mesenchymal stem cells derived from bonemarrow. Cell Tissue Res 2010: 342: 205–212.

94. Hui M, Tenenbaum HC. New face of an old enzyme: alka-line phosphatase may contribute to human tissue aging byinducing tissue hardening and calcification. Anat Rec 1998:253: 91–94.

95. Hunter GK, Goldberg HA. Nucleation of hydroxyapatite bybone sialoprotein. Proc Natl Acad Sci USA 1993: 90: 8562–8565.

96. Ikeda T, Nomura S, Yamaguchi A, Suda T, Yoshiki S. In situhybridization of bone matrix proteins in undecalcifiedadult rat bone sections. J Histochem Cytochem 1992: 40:1079–1088.

97. Ikezawa K, Hart CE, Williams DC, Narayanan AS. Charac-terization of cementum derived growth factor as an insu-lin-like growth factor-I like molecule. Connect Tissue Res1997: 36: 309–319.

98. Ivanovski S, Komaki M, Bartold PM, Narayanan AS. Peri-odontal-derived cells attach to cementum attachment pro-tein via alpha 5 beta 1 integrin. J Periodontal Res 1999: 34:154–159.

99. Itoiz ME, Mayo J, Cabrini RL, Carranza FA Jr. Histochemi-cal study of healing wounds: alkaline and acid phospha-tase. J Oral Surg 1969: 27: 641–644.

100. Iwata T, Morotome Y, Tanabe T, Fukae M, Ishikawa I, OidaS. Noggin blocks osteoinductive activity of porcine enamelextracts. J Dent Res 2002: 81: 387–391.

101. Janones DS, Massa LF, Arana-Chavez VE. Immunocyto-chemical examination of the presence of amelogenin dur-ing the root development of rat molars. Arch Oral Biol2005: 50: 527–532.

102. Jono S, Peinado C, Giachelli CM. Phosphorylation of os-teopontin is required for inhibition of vascular smoothmuscle cell calcification. J Biol Chem 2000: 275: 20197–20203.

103. Kagayama M, Li HC, Zhu J, Sasano Y, Hatakeyama Y, Miz-oguchi I. Expression of osteocalcin in cementoblasts form-ing acellular cementum. J Periodontal Res 1997: 32: 273–278.

104. Kaipatur NR, Murshed M, McKee MD. Matrix Gla proteininhibition of tooth mineralization. J Dent Res 2008: 87:839–844.

105. Kalpidis CD, Ruben MP. Treatment of intrabony periodon-tal defects with enamel matrix derivative: a literaturereview. J Periodontol 2002: 73: 1360–1376.

106. Kaufmann B, M€uller S, Hanisch F, Hartmann U, PaulssonM, Maurer P, Zaucke F. Structural variability of BM-40/SPARC/osteonectin glycosylation: implications for colla-gen affinity. Glycobiology 2004: 14: 609–619.

107. Kawase T, Okuda K, Yoshie H, Burns DM. Anti-TGF-betaantibody blocks enamel matrix derivative-induced upreg-ulation of p21WAF1/cip1 and prevents its inhibition ofhuman oral epithelial cell proliferation. J Periodontal Res2002: 37: 255–262.

108. K�emoun P, Laurencin-Dalicieux S, Rue J, Farges JC,Gennero I, Conte-Auriol F, Briand-Mesange F, Gade-lorge M, Arzate H, Narayanan AS, Brunel G, Salles JP.Human dental follicle cells acquire cementoblast fea-tures under stimulation by BMP-2/-7 and enamelmatrix derivatives (EMD) in vitro. Cell Tissue Res 2007:329: 283–294.

109. King GN, King N, Hughes FJ. The effect of root surfacedemineralization on bone morphogenetic pro-tein-2-induced healing of rat periodontal fenestrationdefects. J Periodontol 1998: 69: 561–570.

110. King GN. New regenerative technologies: rationale andpotential for periodontal regeneration: 1. New advances inestablished regenerative strategies. Dent Update 2001: 28:7–12.

111. King GN. New regenerative technologies: rationale andpotential for periodontal regeneration: 2. Growth factors.Dent Update 2001: 28: 60–65.

112. King GN, Cochran DL. Factors that modulate the effectsof bone morphogenetic protein-induced periodontal

Cementum proteins

229

Page 20: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

regeneration: a critical review. J Periodontol 2002: 73:925–936.

113. Komaki M, Iwasaki K, Arzate H, Sampath Narayanan A, Iz-umi Y, Morita I. Cementum protein 1 (CEMP1) induces acementoblastic phenotype and reduces osteoblastic differ-entiation in periodontal ligament cells. J Cell Physiol 2012:227: 649–657.

114. Kuboki Y, Sasaki M, Saito A, Takita H, Kato H. Regenera-tion of periodontal ligament and cementum by BMP-ap-plied tissue engineering. Eur J Oral Sci 1998: 106: 197–203.

115. Kuivaniemi H, Tromp G, Prockop DJ. Mutations in fibrillarcollagens (types I, II, III, and XI), fibril-associated collagen(type IX), and network-forming collagen (type X) cause aspectrum of diseases of bone, cartilage, and blood vessels.HumMutat 1997: 9: 300–315.

116. Lamster IB. Current concepts and future trends for peri-odontal disease and periodontal therapy, Part 2: classifica-tion, diagnosis, and nonsurgical and surgical therapy.Dent Today 2001: 20: 86–91.

117. LaValle P, Daniels K, Hay ED, Olsen BR. Type X collagenis transcriptionally activated and specifically localizedduring sternal cartilage maturation. Matrix 1992: 12:404–413.

118. Liu HW, Yacobi R, Savion N, Narayanan AS, Pitaru S. A col-lagenous cementum-derived attachment protein is a mar-ker for progenitors of the mineralized tissue-forming celllineage of the periodontal ligament. J Bone Miner Res 1997:12: 1691–1699.

119. Luo W, Slavkin HC, Snead ML. Cells from Hertwig’s epi-thelial root sheath do not transcribe amelogenin. J Peri-odontal Res 1991: 26: 42–47.

120. Lynch SE, de Castilla GR, Williams RC, Kiritsy CP, HowellTH, Reddy MS, Antoniades HN. The effects of short-termapplication of a combination of platelet-derived and insu-lin-like growth factors on periodontal wound healing. J Pe-riodontol 1991: 62: 458–467.

121. MacNeil RL, Somerman MJ. Molecular factors regulatingdevelopment and regeneration of cementum. J Periodon-tal Res 1993: 28: 550–559.

122. MacNeil RL, Sheng N, Strayhorn C, Fisher LW, Somer-man MJ. Bone sialoprotein is localized to the root sur-face during cementogenesis. J Bone Miner Res 1994: 9:1597–1606.

123. MacNeil RL, Berry J, D’Errico J, Strayhorn C, Piotrowski B,Somerman MJ. Role of two mineral-associated adhesionmolecules, osteopontin and bone sialoprotein, during ce-mentogenesis. Connect Tissue Res 1995: 33: 1–7.

124. Mar�ıkov�a Z, Kindlov�a M. Akaline phosphatase in the peri-odontium of rat molars. J Dent Res 1972: 51: 1502.

125. Mariotti A. Efficacy of chemical root surface modifiers inthe treatment of periodontal disease. A systematic review.Ann Periodontol 2003: 8: 205–226.

126. Mark MP, Butler WT, Prince CW, Finkelman RD, Ruch JV.Developmental expression of 44-kDa bone phosphopro-tein (osteopontin) and bone y-carboxyglutamic acid(Gla)-containing protein (osteocalcin) in calcifying tissuesof rat. Differentiation 1988: 37: 123–136.

127. Matsuda N, Lin WL, Kumar NM, Cho MI, Genco RJ.Mitogenic, chemotactic, and synthetic responses ofrat periodontal ligament fibroblastic cells to poly-peptide growth factors in vitro. J Periodontol 1992:63: 515–525.

128. McAllister B, Narayanan AS, Miki Y, Page RC. Isolation of afibroblast attachment protein from cementum. J Periodon-tal Res 1990: 25: 99–105.

129. McCauley LK, Nohutcu RM. Mediators of periodontalosseous destruction and remodeling: principles and impli-cations for diagnosis and therapy. J Periodontol 2002: 73:1377–1391.

130. McCulloch CA, Nemeth E, Lowenberg B, Melcher AH.Paravascular cells in endosteal spaces of alveolar bonecontribute to periodontal ligament cell populations. AnatRec 1987: 219: 233–242.

131. McLaren LA, Wildeman AG. Fibronectin receptors in pre-implantation development: cloning, expression and locali-zation of the a5 and b1 inetgrin subunits in bovinetrophoblast. Biol Reprod 1995: 53: 153–165.

132. Melcher AH. On the repair potential of periodontal tissues.J Periodontol 1976: 47: 256–260.

133. Melcher AH. Does the developmental origin of cementum,periodontal ligament and bone predetermine their behav-ior in adults? In: Guggenheim B, editor. PeriodontologyToday. Proceddings of the conference held in Zurich, Swit-zerland, May 6–8. Basel, Switzerland: S. Karger AG, 1988:6–14.

134. Menanteau J, Neuman WF, Neuman MW. A study of boneproteins which can prevent hydroxyapatite formation.Metab Bone Dis Relat Res 1982: 4: 157–162.

135. Merry K, Dodds R, Littlewood A, Gowen M.. Expressionof osteopontin mRNA by osteoclasts and osteoblasts inmodeling adult human bone. J Cell Sci 1993: 104: 1013–1020.

136. Metzger Z, Weinstock B, Dotan M, Narayanan AS, Pitaru S.Differential chemotactic effect of cementum attachmentprotein on periodontal cells. J Periodontal Res 1998: 33:126–129.

137. Miki Y, Narayanan AS, Page RC. Mitogenic activity ofcementum components to gingival fibroblasts. J Dent Res1987: 66: 1399–1403.

138. Miller PD, Craddock RD. Surgical advances in the coverageof exposed roots. Curr Opin Periodontol 1996: 3: 103–108.

139. Mizutani S, Tsuboi T, Tazoe M, Koshihara Y, Goto S, To-gari A. Involvement of FGF-2 in the action of Emdogain onnormal human osteoblastic activity. Oral Dis 2003: 9: 210–217.

140. Muir H. The chondrocyte, architect of cartilage. Biome-chanics, structure, function and molecular biology of car-tilage matrix macromolecules. BioEssays 1995: 17: 1039–1048.

141. Murakami Y, Kojima T, Nagasawa T, Kobayashi H, Ishika-wa I. Novel isolation of alkaline phosphatase-positive sub-population from periodontal ligament fibroblasts. JPeriodontol 2003: 74: 780–786.

142. Murphy KG, Gunsolley JC. Guided tissue regeneration forthe treatment of periodontal intrabony and furcationdefects. A systematic review. Ann Periodontol 2003: 8: 266–302.

143. Nagano T, Oida S, Suzuki S, Iwata T, Yamakoshi Y, OgataY, Gomi K, Arai T, Fukae M. Porcine enamel protein frac-tions contain transforming growth factor-beta1. J Period-ontol 2006: 77: 1688–1694.

144. Nakae H, Narayanan AS, Raines E, Page RC. Isolation andpartial characterization of mitogenic factors from cemen-tum. Biochemistry 1991: 30: 7047–7052.

Arzate et al.

230

Page 21: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

145. Nakagawa E, Zhang L, Shin JO, Kim EJ, Cho SW, OhshimaH, Chen Z, Jung HS. The novel expression of Oct3/4 andBmi1 in the root development of mouse molars. Cell Tis-sue Res 2012: 347: 479–484.

146. Nakashima M, Reddi AH. The application of bone mor-phogenetic proteins to dental tissue engineering. Nat Bio-technol 2003: 21: 1025–1032.

147. Nam H, Kim J, Park J, Park JC, Kim JW, Seo BM, Lee JC,Lee G. Expression profile of the stem cell markers inhuman Hertwig’s epithelial root sheath/Epithelial rests ofMalassez cells.Mol Cells 2011: 31: 355–360.

148. Nanci A. Content and distribution of noncollagenousmatrix proteins in bone and cementum: relationship tospeed of formation and collagen packing density. J StructBiol 1999: 126: 256–269.

149. Narayanan AS, Ikezawa K, Wu D, Pitaru S. Cementum spe-cific components which influence periodontal connectivetissue cells. Connect Tissue Res 1995: 33: 19–21.

150. Nociti FH Jr, Berry JE, Foster BL, Gurley KA, Kingsley DM,Takata T, Miyauchi M, Somerman MJ. Cementum: a phos-phate-sensitive tissue. J Dent Res 2002: 81: 817–821.

151. Nojima N, Kobayashi M, Shionome M, Takahashi N, SudaT, Hasegawa K. Fibroblastic cells derived from bovine peri-odontal ligaments have the phenotypes of osteoblasts. JPeriodontal Res 1990: 25: 179–185.

152. Nu~nez J, Sanz M, Hoz-Rodr�ıguez L, Zeichner-David M,Arzate H. Human cementoblasts express enamel-associ-ated molecules in vitro and in vivo. J Periodontal Res 2010:45: 809–814.

153. Nu~nez J, Sanz-Blasco S, Vignoletti F, Mu~noz F, Arzate H,Villalobos C, Nu~nez L, Caffesse RG, Sanz M. Periodontalregeneration following implantation of cementum andperiodontal ligament-derived cells. J Periodontal Res 2012:47: 33–44.

154. Oda S, Kinoshita A, Higuchi T, Shizuya T, Ishikawa I.Ectopic bone formation by biphasic calcium phosphate(BCP) combined with recombinant human bonemorphogenetic protein-2 (rhBMP-2). J Med Dent Sci1997: 44: 53–62.

155. Oida S, Nagano T, Yamakoshi Y, Ando H, Yamada M, Fu-kae M. Amelogenin gene expression in porcine odonto-blasts. J Dent Res 2002: 81: 103–108.

156. Oldberg A, Franzen A, Heinegard D. The primary structureof a cell-binding bone sialoprotein. J Biol Chem 1988: 263:19430–19432.

157. Olson S, Arzate H, Narayanan AS, Page RC. Cell attach-ment activity of cementum proteins and mechanism ofendotoxin inhibition. J Dent Res 1991: 70: 1272–1277.

158. Oringer RJ. Biological mediators for periodontal and boneregeneration. Compend Contin Educ Dent 2002: 23: 501–514.

159. Oyama M, Suzuki N, Yamaguchi Y, Maeno M, Otzuka K,Ito K. Effect of enamel matrix derivative on the differentia-tion of C2C12 cells. J Periodontol 2002: 73: 543–550.

160. Page RC, Baab DA. A new look at the etiology and patho-genesis of early-onset periodontitis. Cementopathia revis-ited. J Periodontol 1985: 56: 748–751.

161. Pan K, Sun Q, Zhang J, Ge S, Li S, Zhao Y, Yang P. Multilin-eage differentiation of dental follicle cells and the roles ofRunx2 over-expression in enhancing osteoblast/cemento-blast-related gene expression in dental follicle cells. CellProlif 2010: 43: 219–228.

162. Papagerakis P, Macdougall M, Hotton D, Bailleul-ForestierI, Oboeuf M, Berdal A. Expression of amelogenin in odon-toblasts. Bone 2003: 32: 228–240.

163. Park JB, Matsuura M, Han KY, Norderyd O, Lin WL, GencoRJ, Cho MI. Periodontal regeneration in class III furcationdefects of beagle dogs using guided tissue regenerativetherapy with platelet-derived growth factor. J Periodontol1995: 66: 462–477.

164. Paula-Silva FW, Ghosh A, Arzate H, Kapila S, da Silva LA,Kapila YL. Calcium hydroxide promotes cementogenesisand induces cementoblastic differentiation of mesenchy-mal periodontal ligament cells in a CEMP1- andERK-dependent manner. Calcif Tissue Int 2010: 87: 144–157.

165. Pearson CH, Gibson CJ. Proteoglycans of bovine periodon-tal ligament and skin. Occurrence of different hybrid-sul-phated galactosaminoglycans in distinct proteoglycans.Biochem J 1982: 201: 27–37.

166. Petersson U, Hultenby K, Wendel M. Identification, distri-bution and expression of osteoadherin during tooth for-mation. Eur J Oral Sci 2003: 111: 128–136.

167. Pitaru S, Hekmati M, Soldinger M, Savion N. Growth andmigration of gingival epithelial cells on mineralized andpartially demineralized root surfaces in an in vitro system.J Periodontol 1988: 59: 531–534.

168. Pitaru S, Savion N, Hekmati H, Olson S, Narayanan AS.Binding of a cementum attachment protein to extracellu-lar matrix components and to dental surfaces. J Periodon-tal Res 1992: 27: 640–646.

169. Pitaru S, Savion N, Hekmati H, Olson S, Narayanan AS.Molecular and cellular interactions of a cementum attach-ment protein with periodontal cells and cementum matrixcomponents. J Periodontal Res 1993: 28: 560–562.

170. Pitaru S, McCulloch CA, Narayanan AS. Cellular originsand differentiation control mechanisms during periodon-tal development and wound healing. J Periodontal Res1994: 29: 81–94.

171. Pitaru S, Narayanan SA, Olson S, Savion N, Hekmati H, AltI, Metzger Z. Specific cementum attachment proteinenhances selectively the attachment and migration ofperiodontal cells to root surfaces. J Periodontal Res 1995:30: 360–368.

172. Price PA, Urist MR, Otawara Y. Matrix Gla protein, a newgamma-carboxyglutamic acid-containing protein which isassociated with the organic matrix of bone. Biochem Bio-phys Res Commun 1983: 117: 765–771.

173. Price PA. Gla-containing proteins of bone. Connect TissueRes 1989: 21: 51–57.

174. Reichert T, St€orkel S, Becker K, Fisher LW. The role ofosteonectin in human tooth development: an immunohis-tological study. Calcif Tissue Int 1992: 50: 468–472.

175. Reynolds MA, Aichelmann-Reidy ME, Branch-Mays GL,Gunsolley JC. The efficacy of bone replacement grafts inthe treatment of periodontal osseous defects. A systematicreview. Ann Periodontol 2003: 8: 227–265.

176. Ripamonti U, Reddi AH. Periodontal regeneration: poten-tial role of bone morphogenetic proteins. J Periodontal Res1994: 29: 225–235.

177. Ripamonti U, Heliotis M, Rueger DC, Sampath TK. Induc-tion of cementogenesis by recombinant human osteo-genic protein-1 (hop-1/bmp-7) in the baboon (Papioursinus). Arch Oral Biol 1996: 41: 121–126.

Cementum proteins

231

Page 22: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

178. Ripamonti U. Soluble osteogenic molecular signals andthe induction of bone formation. Biomaterials 2006: 27:807–822.

179. Roach HI. Why does bone matrix contain non-collage-nous proteins? The possible roles of osteocalcin, osteo-nectin, osteopontin and bonesialoprotein in bonemineralization and resorption. Cell Biol Int 1994: 18:617–628.

180. Rodrigues TL, Nagatomo KJ, Foster BL, Nociti FH, Somer-man MJ. Modulation of phosphate/pyrophosphate metab-olism to regenerate the periodontium. A novel in vivoapproach. J Periodontol 2011: 82: 1757–1766.

181. Rutherford RB, Ryan ME, Kennedy JE, Tucker MM, Cha-rette MF. Platelet-derived growth factor and dexametha-sone combined with a collagen matrix induceregeneration of the periodontium in monkeys. J Clin Peri-odontol 1993: 20: 537–544.

182. Saito M, Narayanan AS. Signaling reactions induced inhuman fibroblasts during adhesion to cementum-derivedattachment protein. J Bone Miner Res 1999: 14: 65–72.

183. Saygin NE, Giannobile WV, Somerman MJ. Molecular andcell biology of cementum. Periodontol 2000 2000: 24: 73–98.

184. Schild C, Beyeler M, Lang NP, Trueb B. Cementum attach-ment protein/protein-tyrosine phosphatase-like memberA is not expressed in teeth. Int J Mol Med 2009: 23: 293–296.

185. Sch€onfeld HJ, P€oschl B, Wessner B, Kistler A. Altered dif-ferentiation of limb bud cells by transforming growth fac-tors-beta isolated from bone matrix and from platelets.Bone Miner 1991: 13: 171–189.

186. Schroeder HE. Handbook of Microscopy Anatomy, Vol. 5:The Periodontium. Oksche A, Vollrath L, editors.Springer-Verlag, 1986.

187. Schroeder HE. Biological problems of regenerative ce-mentogenesis: synthesis and attachment of collagenousmatrices on growing and established root surfaces. Int RevCytol 1992: 142: 1–59.

188. Schwartz Z, Carnes DL Jr, Pulliam R, Lohmann CH, SylviaVL, Liu Y, Dean DD, Cochran DL, Boyan BD. Porcine fetalenamel matrix derivative stimulates proliferation but notdifferentiation of pre-osteoblastic 2T9 cells, inhibits prolif-eration and stimulates differentiation of osteoblast-likeMG63 cells, and increases proliferation and differentiationof normal human osteoblast NHOst cells. J Periodontol2000: 71: 1287–1296.

189. Scott JE. Collagen-proteoglycan interactions. Localisationof proteoglycans in tendon by electromicroscopy. BiochemJ 1980: 187: 887–891.

190. Scott JE, Orford R. Dermatan sulphate-rich proteoglycanassociates with rat tail-tendon collagen at the d band inthe gap region. Biochem J 1981: 197: 213–216.

191. Selvig KA, Wikesj€o UM, Bogle GC, Finkelman RD.Impaired early bone formation in periodontal fenestra-tion defects in dogs following application of insulin-likegrowth factor (II). Basic fibroblast growth factor andtransforming growth factor beta 1. J Clin Periodontol1994: 21: 380–385.

192. Seo BM, Miura M, Gronthos S, Bartold PM, Batouli S, Bra-him J, Young M, Robey PG, Wang CY, Shi S. Investigationof multipotent postnatal stem cells from human periodon-tal ligament. Lancet 2004: 364: 149–155.

193. Shen G. The role of type X collagen in facilitating and regu-lating endochondral ossification of articular cartilage. Or-thod Craniofac Res 2005: 8: 11–17.

194. Shinmura Y, Tsuchiya S, Hata K, Honda MJ. Quiescent epi-thelial cell rests of Malassez can differentiate into amelo-blast-like cells. J Cell Physiol 2008: 217: 728–738.

195. Shu R, Liu Z, Ge L. Influences of porcine enamel matrixproteins on MC3T3-E1 osteoblast proliferation and differ-entiation. Hua Xi Kou Qiang Yi Xue Za Zhi 2000: 18: 226–228.

196. Slavkin HC, Boyde A. Cementum: an epithelial secretoryproduct? J Dent Res 1974: 53: 157.

197. Slavkin HC, Bessem C, Fincham AG, Bringas P Jr, Santos V,Snead ML, Zeichner-David M. Human and mouse cemen-tum proteins immunologically related to enamel proteins.Biochim Biophys Acta 1989: 991: 12–18.

198. Slavkin HC, Bringas P Jr, Bessem C, Santos V, NakamuraM, Hsu MY, Snead ML, Zeichner-David M, Fincham AG.Hertwig’s epithelial root sheath differentiation and initialcementum and bone formation during long-term organculture of mouse mandibular first molars using serumless,chemically-defined medium. J Periodontal Res 1989: 24:28–40.

199. Sodek J, Ganss B, McKee MD. Osteopontin. Crit Rev OralBiol Med 2000: 11: 279–303.

200. Somerman MJ, Archer SY, Hassell TM, Shteyer A, FosterRA. Enhancement by extracts of mineralized tissues ofprotein production by human gingival fibroblasts in vitro.Arch Oral Biol 1987: 32: 879–883.

201. Somerman MJ, Sauk JJ, Foster RA, Norris K, Dickerson K,Argraves WS. Cell attachment activity of cementum: bonesialoprotein II identified in cementum. J Periodontal Res1991: 26: 10–16.

202. Sonoyama W, Seo BM, Yamaza T, Shi S. Human Hertwig’sepithelial root sheath cells play crucial roles in cementumformation. J Dent Res 2007: 86: 594–599.

203. Spouge JD. A new look at the rests of Malassez. A review oftheir embryological origin, anatomy, and possible role inperiodontal health and disease. J Periodontol 1980: 51:437–444.

204. Techawattanawisal W, Nakahama K, Komaki M, Abe M,Takagi Y, Morita I. Isolation of multipotent stem cells fromadult rat periodontal ligament by neurosphere-formingculture system. Biochem Biophys Res Commun 2007: 357:917–923.

205. Tenorio D, Foyle DM, Hughes FJ. The modulatory role ofcementum matrix on osteoblastic cells in vitro. J Periodon-tal Res 1997: 32: 362–374.

206. Terkeltaub RA. Inorganic pyrophosphate generation anddisposition in pathophysiology. Am J Physiol Cell Physiol2001: 281: C1–C11.

207. Thomas HF. Root formation. Int J Dev Biol 1995: 39: 231–237.

208. Tompkins K, Veis A. Polypeptides translated from alterna-tively spliced transcripts of the amelogenin gene, devoidof the exon 6a, b, c region, have specific effects on toothgerm development in culture. Connect Tissue Res 2002: 43:224–231.

209. Torres-Quintana MA, L�ecolle S, Goldberg M. Effects of ino-sitol hexasulphate, a casein kinase inhibitor, on dentinephosphorylated proteins in organ culture of mouse toothgerms. Arch Oral Biol 1998: 43: 597–610.

Arzate et al.

232

Page 23: Cementum proteins: role in cementogenesis ...s9ab564d99fcaaf1d.jimcontent.com/download/version... · Cementum proteins: role in cementogenesis, biomineralization, periodontium formation

210. T€oz€um TF, Demiralp B. Platelet-rich plasma: a promis-ing innovation in dentistry. J Can Dent Assoc 2003: 69:664.

211. Tsonis PA, Del Rio-Tsonis K, Millan JL, Wheelock MJ.Expression of N-cadherin and alkaline phosphatase inchick limb bud mesenchymal cells: regulation by 1,25-di-hydroxyvitamin D3 or TGF-beta 1. Exp Cell Res 1994: 213:433–437.

212. Vald�es A, Hoz L, Arzate H, Rose T, Narayanan AS. Cemen-tum attachment protein is splice-variant of protein tyro-sine phosphatase-like (proline instead of catalytic Arginine(PTPLA). J Dent Res 2012: 91: 203–209.

213. Van den Bos T, Beertsen W. Alkaline phosphatase activityin human periodontal ligament: age effect and relation tocementum growth rate. J Periodontal Res 1999: 34: 1–6.

214. Van den Bos T, Handoko G, Niehof A, Ryan LM, CoburnSP, Whyte MP, Beertsen W. Cementum and dentin in hyp-ophosphatasia. J Dent Res 2005: 84: 1021–1025.

215. Veis A, Tompkins K, Alvares K, Wei K, Wang L, Wang XS,Brownell AG, Jengh SM, Healy KE. Specific amelogeningene splice products have signaling effects on cells in cul-ture and in implants Bone. J Biol Chem 2000: 275: 41263–41272.

216. Veis A. Amelogenin gene splice products: potential signal-ing molecules. Cell Mol Life Sci 2003: 60: 38–55.

217. Vidal BC, Mello MLS, Valdrighi L. Histochemical and an-isotropical aspects of the rat cementum. Acta Anat 1974:89: 546–559.

218. Villarreal-Ram�ırez E, Moreno A, Mas-Oliva J,Ch�avez-Pacheco JL, Narayanan AS, Gil-Chavarr�ıa I, Zeich-ner-David M, Arzate H. Characterization of recombinanthuman cementum protein 1 (hrCEMP1): primary role inbiomineralization. Biochem Biophys Res Commun 2009:384: 49–54.

219. Viswanathan HL, Berry JE, Foster BL, Gibson CW, Li Y, Ku-lkarni AB, Snead ML, Somerman MJ. Amelogenin: a poten-tial regulator of cementum-associated genes. J Periodontol2003: 74: 1423–1431.

220. Vogel KG, Paulsson M, Heinegard D. Specific inhibition oftype I and type II collagen fibrillogenesis by the small pro-teoglycan of tendon. Biochem J 1985: 223: 587–597.

221. Vukicevic S, Luyten FP, Reddi AH. Stimulation of theexpression of osteogenic and chondrogenic phenotypes invitro by osteogenin. Proc Natl Acad Sci U S A 1989: 86:8793–8797.

222. Wang HL, Pappert TD, Castelli WA, Chiego DJ Jr, ShyrY, Smith BA. The effect of platelet-derived growth fac-tor on the cellular response of the periodontium: anautoradiographic study on dogs. J Periodontol 1994: 65:429–436.

223. Wikesj€o UM, Sorensen RG, Kinoshita A, Jian Li X, WozneyJM. Periodontal repair in dogs: effect of recombinanthuman bone morphogenetic protein-12 (rhBMP-12) onregeneration of alveolar bone and periodontal attachment.J Clin Periodontol 2004: 31: 662–670.

224. Worapamorn W, Xiao Y, Li H, Young WG, Bartold PM. Dif-ferential expression and distribution of syndecan-1 and -2in periodontal wound healing of the rat. J Periodontal Res2002: 37: 293–299.

225. Wu D, Ikezawa K, Parker T, Saito M, Narayanan AS. Char-acterization of a collagenous cementum-derived attach-ment protein. J Bone Miner Res 1996: 11: 686–692.

226. Wuthier RF. A review of the primary mechanism of endo-chondral calcification with special emphasis on the role ofcells, mitochondria and matrix vesicles. Clin Orthop 1982:169: 219–242.

227. Xie Y, Song Z, Shu R, Sun Y, Luo M, Zhang XL. Effects ofcementum attachment proteins on the proliferation andmineralization of monkey bone marrow stromal cells in vi-tro. J Chin Stomatol Res 2008: 2: 14–18.

228. Xiong J, Mrozik K, Gronthos S, Bartold PM. Epithelial cellrests of Malassez contain unique stem cell populationscapable of undergoing epithelial-mesenchymal transition.Stem Cells Dev 2012: 11: 2012–2025.

229. Xu J, Wang W, Kapila Y, Lotz J, Kapila S. Multiple differen-tiation capacity of STRO-1+/CD146+ PDL mesenchymalprogenitor cells. Stem Cells Dev 2009: 18: 487–496.

230. Yamamoto T, Domon T, Takahashi S, Arambawatta AK,Wakita M. Immunolocation of proteoglycans and bone-re-lated noncollagenous glycoproteins in developing acellularcementum of rat molars. Cell Tissue Res 2004: 317: 299–312.

231. Yamashita Y, Sato M, Noguchi T. Alkaline phosphatase inthe periodontal ligament of the rabbit and macaque mon-key. Arch Oral Biol 1987: 32: 677–678.

232. Yokokoji T, Narayanan AS. Role of D1 and E cyclins in cellcycle progression of human fibroblasts adhering tocementum attachment protein. J Bone Miner Res 2001: 16:1062–1067.

233. Yoneda S, Itoh D, Kuroda S, Kondo H, Umezawa A, OhyaK, Ohyama T, Kasugai S. The effects of enamel matrixderivative (EMD) on osteoblastic cells in culture and boneregeneration in a rat skull defect. J Periodontal Res 2003:38: 333–342.

234. Yonemura K, Narayanan AS, Miki Y, Page RC, Okada H.Isolation and partial characterization of a growth factorfrom human cementum. Bone Miner 1992: 18: 187–198.

235. Yuan ZA, Chen E, Gibson CW. Model system for evaluationof alternative splicing: exon skipping. DNA Cell Biol 2001:20: 807–813.

236. Zeichner-David M, Hall F, Williams R, Thiemann F, Yen S,MacDougall M, Slavkin HC. Characterization of proteinkinases involved in dentinogenesis. Connect Tissue Res1995: 33: 87–95.

237. Zeichner-David M. Is there more to enamel matrix pro-teins than biomineralization? Matrix Biol 2001: 20: 307–316.

238. Zeichner-David M, Oishi K, Su Z, Zakartchenko V, ChenLS, Arzate H, Bringas P Jr. Role of Hertwig’s epithelial rootsheath cells in tooth root development. Dev Dyn 2003:228: 651–663.

239. Zeichner-David M, Chen LS, Hsu Z, Reyna J, Caton J, Brin-gas P. Amelogenin and ameloblastin show growth-factorlike activity in periodontal ligament cells. Eur J Oral Sci2006: 114: 244–253.

240. Zeichner-David M. Regeneration of periodontal tissues:cementogenesis revisited. Periodontol 2000 2006: 41: 196–217.

Cementum proteins

233