Sie sind auf Seite 1von 18

Designing Biomaterials for in Situ Periodontal Tissue Regeneration

Hai-Hua Sun and Tie-Jun Qu


Dept. of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, Xian, Shaanxi, P.R. China 710032

Xian-Hua Zhang
Dept. of Stomatology, Periodontology Unit, Chinese PLA General Hospital, Fu-Xing Road, Beijing 100853, China

Qing Yu
Dept. of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, Xian, Shaanxi, P.R. China 710032

Fa-Ming Chen
Dept. of Periodontology & Oral Medicine, Translational Research Team, School of Stomatology, Fourth Military Medical University, Xian, Shaanxi, P.R. China 710032 DOI 10.1002/btpr.698 Published online September 12, 2011 in Wiley Online Library (wileyonlinelibrary.com).

The regeneration of periodontal tissue poses a signicant challenge to biomaterial scientists, tissue engineers and periodontal clinicians. Recent advances in this eld have shifted the focus from the attempt to recreate tissue replacements/constructs ex vivo to the development of biofunctionalized biomaterials that incorporate and release regulatory signals in a precise and near-physiological fashion to achieve in situ regeneration. The molecular and physical information coded within the biomaterials dene a local biochemical and mechanical niche with complex and dynamic regulation that establishes key interactions with host endogenous cells and, hence, may help to unlock latent regenerative pathways in the body by instructing cell homing and regulating cell proliferation/differentiation. In the future, these innovative principles and biomaterial devices promise to have a profound impact on periodontal reconstructive therapy and are also likely to reconcile the clinical and commerC 2011 American Institute of Chemical cial pressures on other tissue engineering endeavors. V Engineers Biotechnol. Prog., 28: 320, 2012 Keywords: cell homing, endogenous regeneration, tissue engineering, controlled release, regenerative medicine, biofunctionalized biomaterials

Introduction
The periodontium is continually undergoing alterations and physiological adjustments due to the effects of aging, occlusal force and other detrimental changes in the oral environment.1 Particularly, the presence of plaque-induced periodontitis results in destruction and pathological changes to the periodontium, i.e., the loss of alveolar bone, periodontal ligament (PDL) and cementum, that unfortunately show very little tendency for self-repair. Hence, once periodontitis becomes established, only effective therapeutic intervention has the potential to induce substantial periodontal tissue regeneration. To date, however, there has been no appropriate method that is available for clinical choice.2 New insights into the reparative capability of the periodontium in conjunction with progress in stem cell biology, molecular science and biomaterials design will enable us to develop novel therapies that use engineered biological compounds and stem cells for therapeutic periodontal regeneration.

H.-H. Sun, T.-J. Qu and X.-H Zhang contributed equally to this work. Correspondence concerning this article should be addressed to Q. Yu at yuqing@fmmu.edu.cn. and F.-M. Chen at cfmsunhh@fmmu.edu.cn.
C 2011 American Institute of Chemical Engineers V

Since Seo et al.3 successfully isolated and characterized PDL stem cells from human PDL tissue in 2004, the use of stem cell delivery/transplantation has become the predominant research strategy in periodontal regenerative medicine.412 Experimental results obtained using various animal models have shown that under certain conditions, PDL stem cells,46 bone marrow mesenchymal stem cells (MSCs),710 adipose-derived stem cells11 and other induced pluripotent stem (iPS) cells12 can potentially induce the formation of new periodontal tissue (Table 1). However, stem cell transplantation requires complicated and specic in vitro cell culture conditions and complex operating procedures. Its clinical translation therefore faces enormous technical and nancial hurdles, especially in the treatment of non-fatal diseases, such as periodontitis.13 Although stem cell therapy will undoubtedly lead to new clinical technologies in the future and warrants further basic research and translational investigation, it would be more advantageous if welldesigned biomedical devices can be applied to achieve in situ tissue regeneration without the use of exogenously manipulated cells. With the progress of stem cell biology research and recombinant protein biotechnology, a promising approach that has recently been proposed is the use of stem
3

Biotechnol. Prog., 2012, Vol. 28, No. 1

Table 1. The Use of Stem Cells as Exogenous Regenerative Tools for Periodontal Tissue Regeneration: Selected Examples and Their Brief Outcomes in Animal Models Stem Cells PDLSCs Vehicles HA/TCP Animal Model Periodontitis models in miniature pigs Brief Outcomes PDLSCs were capable of regenerating periodontal tissues and statistical analysis indicated that both the autologous and allogeneic PDLSCs treatments signicantly improved periodontal tissue regeneration compared with control groups, with no difference between the autologous and allogeneic PDLSCs groups Periodontal regeneration was signicantly observed with both newly formed cementum and well-oriented PDL bers more in the PDL cell group than in the iliac bone marrow mesenchymal stromal cell and alveolar periosteal cell groups Four weeks after transplantation, the periodontal defects were almost regenerated with periodontal tissue and transplanted BM-MSCs could survive and differentiate into periodontal tissue-forming cells Both freshly isolated and cryopreserved BM-MSCs induced signicantly better periodontal regeneration with newly formed cementum, alveolar bone and PDL compared with the application of collagen scaffold alone Implantation of BM-MSCs regenerated signicantly greater new bone and more appropriately orientated PDL bers; and donor-derived BM-MSCs were found integrated in newly formed bone, cementum and PDL A small amount of alveolar bone regeneration was observed 2 and 4 weeks after ASC/PRP implantation. Moreover, 8 weeks after implantation, a PDL-like structure was observed along with alveolar bone Histological analysis showed much more new alveolar bone and cementum formation (with regenerated PDL between them) in iPS cell group compared with both control groups (cellfree groups) Ref.(s) 4,5

Cell sheets with b-TCP/collagen scaffold

One-wall intrabony defects in dogs

BM-MSCs

Atelocollagen (2% type I collagen) Collagen scaffold

Experimental Class III periodontal defects in beagle dogs

7,8

Periodontal fenestration defects (5 5 mm) in beagle dogs

Gelatin beads

Periodontal tissue defect in rats (surgically created on the buccal of the mandibular rst molar by removing bone, PDL and cementum) Periodontal tissue defect in rats

10

ASCs

PRP

11

iPS cells

Silk scaffold EMD

Critical-size calvarial bone defects in mice

12

ASCs, adipose-derived stem cells; BM-MSCs, bone marrow mesenchymal stromal cells; EMD, enamel matrix derivatives; HA, hydroxyapatite; iPS cells, induced pluripotent stem cells; PDL, periodontal ligament; PDLSCs, periodontal ligament stem cells; PRP, platelet-rich plasma; TCP, tricalcium phosphate.

cell migration mechanisms in the body to develop new biofunctionalized biomaterials that actively recruit a patients own stem cells to a desired location for therapeutics (Figure 1). The promotion of in situ periodontal regeneration is expected to ameliorate the translational difculties in relation to the use of exogenous stem cells and may have broad prospects and potential for routine clinical applications.1315 In recent years, Mao and coworkers have put this principle into practice by using biomaterials complexed with cell homing factors to recruit the bodys own stem cells in vivo.1618 When an incisor scaffold was implanted orthotopically following mandibular incisor extraction in rats, a putative PDL and new bone regenerated at the interface of a rat incisor scaffold with native alveolar bone provided that stromalderived factor (SDF)-1 and bone morphogenetic protein (BMP)-7 were delivered to scaffold microchannels.16 Similarly, a chemotaxis-based approach has demonstrated the capability of recellularization and revascularization in endodontically treated root canals in vivo, although in an ectopic model.17 Furthermore, recent data from the same group suggest that the entire articular surface of the synovial joint can be regenerated by collagen hydrogels that are spatially

infused with transforming growth factor (TGF)-b3 without cell transplantation in a skeletally mature rabbit model in which the entire articular surface of unilateral proximal humeral condyles has been surgically excised.18 Mao and coworkers endeavor is a renaissance of use of the host as a bioreactor and recruitment of the hosts endogenous cells for regeneration. They have bucked the current trend for use of ex vivo cultivated stem cell preparations for tissue engineering. At the same time, other scientists have successfully regulated the cell recruitment that has demonstrated effective and safe for bone and other tissue regeneration (Table 2).1930 However, cell homing strategies are currently in the early stages of development, which require further optimization; moreover, there are hurdles to overcome before seriously discussing the adaptation of current tested paradigms to large animals and human beings. To date, various bioengineering research strategies have sought to induce cell recruitment and direct cell fates toward tissue formation via the incorporation of signaling molecules and the functionalization of scaffolding materials.15 These strategies have become increasingly popular in the eld of stem cell research and regenerative medicine. Given that

Biotechnol. Prog., 2012, Vol. 28, No. 1

Figure 1. Schematic representation of two stem cell migration mechanisms in the body that may be regulated for the recruitment of endogenous cells (illustrations not to scale, modied from Chen et al.15).
(Top) Stem cells within the extracellular matrix (ECM) (or located in the neighboring cell niches of a injury) can recognize and obey directional cues and reach site of injury via interstitial migration or active amoeboid movement, which can occur independent of blood ow. (Bottom) Stem cells from distance cell niches (i.e., the bone marrow) can be mobilized into the blood (i.e., by injury) and disseminated throughout the body until they recognize and interact with microvascular endothelial cells at a particular target disease site. A coordinated multistep process ensues, including stem cell rolling, adhesion to the endothelium, transendothelial migration, chemotaxis, and invasion of the ECM, where the cells replenish and maintain the cell niche and hence enhance the regenerative potential of the tissue or directly reach the site of injury; they then participate in tissue repair and regeneration.

different bioactive factors play different key roles in the different stages of tissue reconstruction, such as cell recruitment, cell proliferation/differentiation and tissue integration/vascularization, one critical issue that must be addressed is the design and development of biomaterials that can provide the controlled spatiotemporal release of various key signaling molecules during different stages of regeneration. An understanding of which factors to deliver and the timing of delivery is emerging from advances in the elucidation of the critical pathways that are involved in the development of integrated periodontal tissues. With modern drug delivery technology, biomaterials can be designed to release a variety of critical exogenous signaling molecules to mobilize/activate the bodys stem cell niches and regulate them toward functional regeneration. This is a new research direction for biomaterials design in tissue engineering and regenerative medicine and has important clinical signicance and the potential to be applied broadly in the near future.1315 This article addresses the design of novel medical devices for periodontal regeneration and highlights the development of the state of the art of biofunctionalized scaffolds that incorporate signaling molecules required to capture host own cells and catalyze endogenous repair. Furthermore, it describes the challenges and future directions in this rapidly advancing eld. Although our focus in this manuscript is on the regeneration of periodontal tissues, the concept should be more generally applicable.

Clinical Need for Periodontal Regeneration


Periodontitis is a worldwide chronic inammatory disease that afict up to 50% of adults worldwide and more than 15% display severe disease concomitant with early the destruction/damage of the periodontium, i.e., the tissues that surround and support the teeth, and if left untreated, can lead to the loosening and subsequent loss of teeth.1 It is also closely related to cardiovascular disease, diabetes and other systemic diseases.31Around the world, the occurrence of periodontal disease results in signicant health and qualityof-life complications.32,33 There may not be any other chronic infectious disease that can affect as many people globally as periodontal disease does. Therefore, it has become an important public health issue.34,35 The ultimate aim of periodontal therapy in perspective is the restoration of lost tissue to its original form and function, i.e., the reformation of an epithelial seal, deposition of new acellular extrinsic ber cementum and insertion of functionally oriented connective tissue bers into the root surface and restoration of alveolar bone height. Current treatment, however, is generally unable to achieve this goal.34 The complete regeneration of the periodontal attachment is a highly desirable outcome of periodontal therapy that is directed at undoing the harms that this disease causes to periodontal structures. Although conventional periodontal treatment (e.g., debridement/scaling and ap surgery) can control further development of periodontal disease and arrest further loss of

6
Table 2. Selected Examples of Recruiting Endogenous Cells for Tissue Regeneration Stimulators/Mediators SDF-1 and BMP-7 Biomaterials Anatomically shaped tooth scaffolds/collagen gel solution Target Cells Not shown

Biotechnol. Prog., 2012, Vol. 28, No. 1

Animal Models/Outcomes SDF-1 and BMP-7 not only recruited signicantly more endogenous cells but also enabled greater angiogenesis than growth-factor-free controls, leading to a putative periodontal ligament and new bone regenerated at the interface of rat incisor scaffold with native alveolar bone Chemotaxis-based approach has potent cell homing effects for recellularization and revascularization in endodontically treated root canals in vivo (an ectopic model in mouse) The entire articular surface of the synovial joint can regenerate without cell transplantation in rabbits Recruited BMOPCs contribute to ectopic bone formation in mice. CTPs are physiologically mobilized into the fracture healing site in the parabiotic mice model

Ref.(s) 16

bFGF, VEGF, or PDGF with a basal set of NGF and BMP-7 TGF-b3

Collagen gel solution

Not shown

17

BMP-2 Endogenously produced factors in response to injury (transverse bular fracture) SDF-1a Endogenously produced EPC-mobilizing cytokines, such as VEGF, SDF-1 and MCP-1 SDF-1a Endogenously produced cytokines (not shown) BMP-2 SDF-1 and BMP-2

An anatomically correct bioscaffold (PCL-HA)/collagen gel solution Collagen pellet

Not shown

18

BMOPCs CTPs

21,22 23

Porous 3D scaffolds made of mineralized collagen

BMSCs EPCs

Efcient invasion of BMSCs into internal compartments of the scaffolds both in vitro and in vivo (a mouse model) Mobilization of EPC population during distraction osteogenesis in human limb-lengthening patients.

24 25

A bioactive knitted silk-collagen sponge scaffold

Fibroblast-like cells BM-derived cells

Recruitment of broblast-like cells for in situ tendon regeneration in a rat Achilles tendon injury model Inammation-induced inux of BM-derived cells into the infarction area in rat myocardial infarction models Recruited BMOPCs contribute to the ectopic de novo generation of bone tissue Synergistic effects of SDF-1 and BMP-2 on the recruitment of osteogenic cells, enhancing both ectopic and in situ bone regeneration in rats

26 27 29 30

Gelatin hydrogels Gelatin hydrogels

BMOPCs BM-derived cells

bFGF, basic broblast growth factor; BM-derived cells, bone marrow-derived cells; BMOPCs, bone marrow-derived osteoblast progenitor cells; BMP, bone morphogenetic protein; BMSCs, bone marrow stromal cells; CTPs, circulating osteogenic connective tissue progenitors; EPCs, endothelial progenitor cells; HA, hydroxyapatite; MCP-1, monocyte chemoattractant protein; NGF, nerve growth factor; PCL, poly-e-caprolactone; PDGF, plateletderived growth factor; SDF-1, stromal cell-derived factor-1; 3D, three-dimensional; TGF, transforming growth factor; VEGF, vascular endothelial growth factor.

periodontal support tissue to a certain extent, it cannot properly regenerate lost periodontal tissue. The wide development and clinical application of bone grafting and guided tissue/bone regeneration techniques have greatly improved the efcacy of clinical periodontal therapy, but the goal of appropriately restoring the lost structure and function of periodontal tissue has not yet been achieved in many, if not all, cases.36,37 Tissue engineering, often used synonymously with regenerative medicine, encompasses a broad range of strategies to replace or regenerate severely damaged or irreversibly affected tissues or organs. In recent years, stem cell biology and tissue engineering research have made rapid advances. The use of stem cell therapy, together with tissue engineering principles, to promote periodontal regeneration has attracted increasing attention and concern and has become the focus of research in this arena.1,34 This requires the exploitation of these cells either by transplanting them from an exogenous source311 or by activating endogenous stem cells pharmacologically.13,15 However, several technical and clinical challenges must be overcome before ex vivo cultured stem cells

can be used to treat patients. This is because there are many bottleneck problems surrounding the clinical translation of stem cell delivery-based therapy that have not yet been resolved.38 Potentially useful cell populations already exist in the body, and attracting these cells to the diseased site (e.g., the damaged periodontium) by designing instructive biomaterials has the potential to provide new therapeutic options for in situ regeneration (Figure 2). The cost and complexity should be reduced in terms of approaches that use ex vivo bioreactors and cell delivery.39 Therefore, the ability to recruit these host cells and to stimulate and enhance the bodys own ability to regenerate lost periodontal tissue has important clinical signicance and the potential to accelerate clinical translation.14

Endogenous Regeneration of Periodontal Tissues


It is in the light of improvements in our understanding of wound repair, concerted research efforts based on the concepts of tissue engineering and regenerative medicine have begun to harness advances in materials science and recombinant DNA technology to yield next generation of

Biotechnol. Prog., 2012, Vol. 28, No. 1

Figure 2. Schematic representation of the use of cell-instructive biomaterials in capturing endogenous cells for tissue regeneration (in vivo tissue engineering strategy).
This schematic assumes that there is already an existing degree of periodontal dysfunction that cannot be restored by endogenous mechanisms alone. The aim of cell-instructive biomaterials is to use biological stimuli and biomimetic scaffolds to harness the bodys innate ability to heal through strategies that avoid in vitro cell manipulation and ex vivo cell delivery. Such biomaterials must be designed to bear complex information inuences (encapsulation of cues) coded within their physical and chemical structures to mimic, as closely as possible, a natural extracellular milieu to recruit/ capture enough endogenous cells from extracellular matrices (ECM) and/or blood and thereafter inuence their behavior in vivo for regeneration of lost/damaged periodontal tissues.

biomaterials-based medical devices for replacing tissue function and new treatment approaches designed to stimulate or augment endogenous repair mechanisms. This development is also true for the regeneration of tooth-supporting tissues that have been lost to periodontal disease.14 Keeping this in mind, an alternative, perhaps simpler, regenerative strategy to periodontal regeneration therefore is to promote endogenous repair/regeneration through design of instructive biomaterials and delivery of growth factors (GFs) and/or other signaling molecules that stimulate/recruit host endogenous cells, including stem/progenitor cells, to invade an injury site and direct robust extracellular matrix (ECM) synthesis and in situ tissue regeneration.

Roles of growth factors Broadly speaking, even without any therapeutic intervention, living tissues can have a staggering capacity for regeneration, normally termed self-repair or endogenous regeneration.15 Endogenous periodontal regeneration requires an orchestrated process involving coordinated migration, proliferation and differentiation of cells from host tissue resources (i.e. the healthy portion of the periodontium or the remaining alveolar bone) specically contributing to restoring the tooth-supporting apparatus. The innate abilities of organization and self-repair unfortunately do not offer a universal regenerative solution.14 Over the past several decades, the discovery of GFs has led to much hope and speculation about the use of these potent peptides to augment the regenerative potential of tissue in the treatment of wounds that are

difcult to heal. In terms of periodontal therapy, studies have shown that the local application of biologically active factors, such as platelet-derived growth factor (PDGF), TGFb, acidic and basic broblast growth factors (a- and bFGF), insulin-like growth factors (IGF-I and -II), cementum-derived growth factor (CGF) and BMPs, can stimulate self-repair and the regeneration of body tissues and can result in the endogenous regeneration of the periodontium.2,14,15,40,41 GFs are found either as matrix bound proteins attached to the ECM or as soluble molecules secreted by cells or cleaved from the matrix by certain enzymes or proteases. They are large polypeptides that modulate cellular activities including proliferation, differentiation, migration, adhesion and gene expression and regulate the mechanisms and pathways that govern wound healing and tissue regeneration.42 It is now well recognized that GFs play crucial roles in the complex cascade of biological events of wound healing, by stimulating chemotaxis and cellular proliferation, by transferring information among cells of the same and different type and their microenvironment, by controlling ECM formation and angiogenesis, by regulating the process of contraction and by re-establishing tissue integrity.4348 GFs initiate their action by binding to specic receptors on the surface of target cells, and their effects are concentration-dependent, often in a complex non-monotonic way.43 The availability of GFs from the conditioned medium of cultured human cells, their expansion through recombinant technologies and improved understanding of their functions and clinical applications has increased the need for pharmaceutical forms.4446 Unfortunately, although many recombinant GFs are now available

Biotechnol. Prog., 2012, Vol. 28, No. 1

for research purposes and some have also been tested in humans, the clinical experience so far has been disappointing.47,48 Because exogenous GFs are expensive, have a short halflife and are quickly diluted and metabolized when used locally or are inactivated through in vivo enzymatic degradation, their conventional routes of administration are unlikely to be effective.42 Concerns and complications associated with burst release of high doses of GFs, rapid degradation and thus low local availability following bolus injection have motivated extensive recent study into sustained delivery strategies that may provide efcacy at lower doses.45,46 From a careful examination of the literature, it is soon realized that only with an appropriate carrier/delivery system can locally applied GFs play a sustained and efcient role in promoting tissue regeneration.39,40,4244 Particularly, it seems that any treatment aimed at mimicking the natural tissue regeneration processes that occur in precise space and time during the development of the periodontium should not be limited to the provision of a single GF but should deliver multiple agents in an optimized ratio and a specic spatiotemporal pattern.39,40,48 Hence, dual/multiple GF delivery is the focus of GF application in periodontal healing via endogenous regeneration.4448 Currently, however, well-established strategies and techniques are lacking with respect to multiple delivery and clinical application. In this regard, the research and development of controlled-release systems of GFs have been and still are important research focuses in periodontal tissue engineering and regenerative medicine.39,40,42

Design of material vehicles For over two decades, a wide range of materials such as natural polymers (e.g., alginate,49 collagen,50 and chitosan51), synthetic polymers (e.g., polylactides, polyglycolides, polyurethanes, and polycaprolactones),5254 ceramics (e.g., silicatebased glasses, calcium sulfate hemihydrate/dihydrate and calcium phosphates)55 and more often their composites5660 have been intensively studied and widely used as delivery vehicles for GFs (Table 3), However, the release of GFs from conventional vehicles is typically governed by molecular diffusion and material degradation, which do not allow for precise and near-physiological delivery of therapeutics to sites of injury. Hence, there is a need to devise more advanced delivery systems to deliver GFs in a controlled, sustained and localized fashion.42 Hydrogels, due to their unique biocompatibility, exible methods of synthesis, range of constituents and desirable physical characteristics, have been one of the most attractive materials of choice for GF delivery applications in regenerative medicine, and myriad hydrogel-based formulations fabricated from natural and synthetic polymers have been proposed for controlled delivery systems.41,42 However, the lack of space provision by hydrogels may be one of the major issues that needs to be addressed in design of material vehicles for tissue engineering applications.58,61 Therefore, a hydrogel-based carrier need to be modied to provide support for new tissue formation and maturation during the more advanced stages of healing. In this regard, Wikesjo et al. (2003)62 proposed a space-providing macroporous expanded polytetrauoroethylene (ePTFE) device to control volume and geometry of rhBMP-2/absorbable collagen sponge (ACS)-induced alveolar bone augmentation.

In terms of cell penetration and tissue ingrowth, the porosity and pore size of most hydrogels may not meet the requirements for tissue engineering scaffolds, which is another critical design consideration for hydrogels in regenerative medicine.61 To develop a hydrogel vehicle with proper porosity and pore size for periodontal regeneration, we have recently designed a macroporous glycidyl methacrylate derivatized dextran (Dex-GMA)/gelatin (Gtn) hydrogel system that may incorporate BMP-loaded microspheres into its porous structures (Figure 3).59 The envelopment of two types of microspheres, each preloaded with different GFs, into the inner pores of Dex-GMA/Gtn hydrogels enables the integration of drug delivery carriers and tissue engineering scaffolds into a single device and, to some extent, allows for the controlled release of dual GFs.63 While signicant work has been dedicated to the development of material vehicles that deliver GFs directly, a relatively simpler approach involves the conversion of cells into protein-producing factories, normally termed gene delivery.6466 However, we must be aware of that material vehicles are also required for in vivo gene delivery. For example, a chitosan/collagen scaffold was used to delivery adenoviruses expressing either BMP-7, PDGF-B, or a combination of both BMP-7 and PDGF-B in surgically induced defects on both sides of the dog mandible.65 This study demonstrates the promising potential of biomaterial expression of combinations of growth factors such as BMP-7 and PDGF-B for bone regeneration in tissue engineering applications. In another impressive investigation, a collagen matrix containing an adenoviral vector encoding PDGF-B demonstrated regenerative and safety capabilities for bone tissue engineering and osseointegration in alveolar bone defects that are comparable with recombinant human PDGF-BB protein delivery in vivo.66 The use of gene delivery for periodontal regeneration is a relatively new strategy because non-viral vectors are continually optimized for clinical therapeutic purposes, and signicant reservations about viral vectors have increasingly dampened their appeal.67 Novel gene delivery systems that act as localized gene depots, providing prolonged maintenance of the therapeutic level of encoded proteins, thus have great potential for future clinical periodontal regeneration.68 Furthermore, we should be aware that novel delivery systems developed for bone or other tissue regeneration can also be applied to periodontal research directly or with slight modication.48,6972

Mobilization of endogenous cells Tissue regeneration requires the presence of a sufcient number of stem cells at the injury site. With the stimulation of local GFs and the support of bioscaffolds, stem cells differentiate into different tissue-forming cells, which is critical to the formation of new tissue.7375 Previous studies have mainly focused on the transplantation of high concentrations of ex vivo cultured pluripotent stem cells. In addition to the low rate of engraftment and other unsolved problems, stem cell transplantation requires in vitro cell manipulation and ex vivo cell delivery, according to strict operating procedures. Furthermore, its clinical application faces enormous technical and nancial challenges. Given the current options for periodontal disease treatment and its non-fatal nature, the benets of conventional stem cell therapy may not outweigh these drawbacks.13,16,17 Therefore, the ability to mobilize the bodys own stem cells and avoid cell culture and

Biotechnol. Prog., 2012, Vol. 28, No. 1


Table 3. Selected Examples of Material Vechicle for Periodontal and Bone Regeneration Growth Factors BMP-2 Biomaterial Carriers Peptide-modied alginate hydrogel Biodegradable collagen hydrogel Animal Model Critically-sized segmental bone defects in rats Class III furcation defects in beagle dogs Conclusion(s) The hybrid alginate/nanober mesh system is a promising growth factor delivery strategy for the repair of challenging bone injuries a topical application of rhBMP2 and rh beta-NGF may improve the quality and quantity of regenerated bone in articially created Class III furcation defects of beagle dogs PDGF-BB-loaded chitosan sponge may be benecial to enhance periodontal bone regeneration DBM and Bio-Oss performed well as carriers for rhBMP-2-driven periodontal regeneration The combined delivery of PDGFBB and IGF-I resulted in signicant increases in periodontal tissue regeneration Suitable application of rhBMP-2 can produce considerable periodontal tissue regeneration, even in cases of horizontal circumferential defects Although both carriers loaded with BMP-2 appear to enhance periodontal regeneration, the renement in carrier systems may provide the key to enhancement of the regenerative potential Characteristics of the PLGA composite makes it an attractive matrix to support native wound healing and rhGDF-5-enhanced periodontal regeneration The PLGA composite appears to be an effective ease-of-use candidate for delivery of growth factors to the periodontium Glycidyl methacrylate derivatized dextran/gelatin microsphere/scaffold system was promising to delivery growth factors for catalyzing periodontal regeneration The present coaxial system was very effective in manufacturing PLGA core-alginate shell microcapsules and in encapsulating multiple biomolecules essential for stem cell differentiation Ref.(s) 49

BMP-2 and b-NGF

50

PDGF-BB BMP-2 PDGF-BB and/or IGF-I BMP-2

Chitosan sponge canine DBM, Bio-Oss, ACS, PLGA or Drilac A methylcellulose gel vehicle

Calvarial defects in rats Routine critical size supra-alveolar periodontal defects in dogs Ligature-induced periodontitis in non-human primates Horizontal circumferential defects created by experimental periodontitis 3-wall intrabony periodontal defects in the baboon

51 52 53

A sponge-type carrier material made of gelatin and polylactic acid polyglycolic acid copolymer ACS or calcium phosphates putty

54

BMP-2

55

GDF-5

Composite matrix constituted by bioresorbable PLGA and additives forming in situ Injectable PLGA composite carrier Dextran and collagen composite hydrogel

Periodontal defects in dogs

56

GDF-5

Bilateral 4 5 mm (width depth) one-wall, critical-size, intra-bony periodontal defects Class III furcation defects in beagle dogs

57

BMP-2

59,61

BMP-2 and dexamethasone

PLGA and alginate

In vitro rat BMSCs culture model

60

ACS, absorbable collagen sponge (type I bovine collagen); Bio-Oss, bovine deorganied crystalline bone matrix; BMP-2, bone morphogenetic protein-2; DBM, demineralized bone matrix; Drilac, polylactic acid granules; GDF-5, growth/differentiation factor-5; IGF-I, insulin-like growth factor I; NGF, nerve growth factor; PDGF-BB, platelet-derived growth factor BB; PLGA, poly(L-lactide-co-glycolide).

transplantation to stimulate self-repair in patients is very promising and may accelerate clinical translation.14,15 Recently, researchers have shown that with the help of GF induction, bone marrow-derived precursor cells in peripheral blood circulation can be recruited for ectopic bone formation.2022,24,25 Kim et al.16,17 have also conrmed that by recruiting the bodys own stem cells it may be possible to achieve a certain degree of odontogenic tissue regeneration. Further exploration of delivery strategies and release criteria for exogenous induction factors in the design of novel functional biomaterials as delivery systems may ultimately lead to endogenous regeneration of targeted tissues.14,15 In addition, the identication of different key factors at different stages of the wound healing cascade (i.e., cell recruitment, cell proliferation/differentiation and tissue remolding and

integration/vascularization) is also very important in the development of safer and more effective approaches of using bioactive factors in the future, which will have far-reaching effects on the repair and regeneration treatment of a variety of tissue defects.1315,39

Crucial Barriers to Progress


Not surprisingly, given the pressing clinical need, the market for biomaterials-based treatments in periodontitis is growing at a rapid rate. Similarly, an emerging philosophy in periodontal regenerative medicine is that rather than attempting to recreate tissue replacements of living periodontium ex vivo, we should aim to develop synthetic materials that establish key interactions with cells in ways that unlock

10

Biotechnol. Prog., 2012, Vol. 28, No. 1

Figure 4. A selected representative scanning electron microscopy image of a freeze-dried particulate vehicle incorporated with soluble growth factors (sample from a large-sized batch of microparticles, as shown in Chen et al.,59 Fig. 2D).

Figure 3. Representative scanning electron microscopy images of two types of glycidyl methacrylate derivatized dextran (Dex-GMA)/gelatin(Gtn) scaffolds developed in our laboratory.
(A) Dex-GMA/Gtn scaffolds with an interconnected pore structure containing microencapsulated growth factors; (B) a particulate-based scaffold obtained from the chemical combination of batches of Dex-GMA/gelatin microspheres loaded with growth factors (unpublished data).

the bodys innate ability to heal.13,14 As described above, endogenous regeneration involves the coordination of a variety of biological signaling molecules. It is the result of in vivo stem cell recruitment, proliferation and differentiation until vascularized tissue forms.75 This process requires a welldesigned functional scaffold that can support cell penetration and tissue ingrowth and a drug delivery system (DDS) that can provide the sequential release of multiple factors to the defect region.7679 Although promising, the current solutions do not feature medical devices that meet general the requirements of clinical application. Barriers in release technology There is increasing evidence that maximizing the potency of tissue regenerative therapies will require design and development of release technology that provides controlled spatiotemporal delivery of multiple key signaling molecules. Richardson et al.80rst reported the use of microspheres and scaffolds to coordinate the release of two GFs in 2001. Since then, many scientists have used different strategies to achieve various GF release proles to meet the needs of different tissue regeneration applications. To date, the simultaneous delivery,63,81 sequential delivery82,83 and 70,84,85 spatiotemporal delivery of different GFs have all been achieved in laboratory studies. Delivery strategies include, but are not limited to, the following86: direct incorporation

(Figure 4), layer-by-layer technology, multiphase loading and particulate-based delivery systems (Figure 3). The introduction of various controlled-release microsphere systems that are preloaded with various bioactive factors into welldesigned scaffolds makes the release of several GFs a possibility (Figure 5).8689 Alongside these positive developments, progress in biomaterial design and drug delivery is converging to produce a new generation of materials for endogenous regeneration.90 To date, however, there is no true GF controlled-release system that has successfully been applied in clinical practice. This is, at least in part, because many difculties and challenges are associated with the delivery of multiple factors, such as the selection and combination of different factors, the establishment of their dose and release criteria and the identication of dose-effect relationships.86 These technical difculties have greatly restricted the application of delivery systems and release technology for the treatment of disease.86,89

Barriers in biomaterial design Biomaterials created today for tissue regeneration are routinely information-rich, and recently, their degree of sophistication has increased signicantly because scaffolding biomaterials must be designed to bear complex information inuences that are coded within their physical and chemical structures to mimic, as closely as possible, a natural extracellular milieu to inuence cell behavior and control cell fate in vivo.90,91 However, for commercialization and clinical use, therapeutic biomaterials must not only be scientically sound and practically efcacious but must also be technically simple and economical; these requirements introduce a potential dichotomy between the need for sophistication and the ease of production.91 The success of a therapeutic material device in the market will nally depend on a combination of its safety, effectiveness, accessibility and cost-effectiveness.91 It is worth mentioning that current major research in the eld of tissue engineering and regeneration focuses on the use of DDS directly or in combination with other biomaterials as cell vehicles for cell transplantation.87,88 The translational difculties faced by cell transplantation, as described previously in this manuscript, also affect the clinical use of DDSs as medical devices. Based on a careful examination of

Biotechnol. Prog., 2012, Vol. 28, No. 1

11

Figure 5. Schematic of the sequential release of multiple growth factors from an assumed particulate-based delivery systems.
Each growth factor is pre-encapsulated into a polymer microsphere system that has unique release kinetics. Sequential release may be achieved by the incorporation of multiple microsphere systems into hydrogel scaffolds.

the literature, it can be understood that cell-recruiting designs based on endogenous regeneration have not received enough attention in existing biomaterials (DDS or scaffold) research and development.1315 The development of cellrecruitment devices to carry exogenous signaling molecules that can stimulate the bodys own repair and regeneration mechanisms after implantation have great potential for clinical application, which may accelerate the application of novel biomaterials to modern clinical practice and hence warrants further explorations.89

Biofunctionalization of Biomaterials
Biomaterials play an essential role in the design of medical devices and delivery systems for periodontal therapy. However, in nearly every case, these materials were adopted from other areas of medical science without substantial redesign for periodontal regenerative use. Further synergism of biologists, materials scientists and periodontists promises to deliver sophisticated biomaterials that display and release multiple selected key signals to meet the physiological requirements of the periodontal tissue repair processes. Those biofunctionalized biomaterials may have a profound impact on periodontal regenerative medicine and will advance clinical translation of tissue engineering-based therapeutics. Cell recruitment design The stem cell homing concept in endogenous tissue regeneration research broadly includes two basic forms of cell recruitment in the body15: (i) stem cells reach the target tissue or organ in a manner similar to hematopoietic stem cells (HSCs) homing to bone marrow after bone marrow transplantation; and (ii) under the inuence of stem cell homing inducers, stem/progenitor cells from the stem cell niche around the tissue damage zone react to a particular microen-

vironment and, through amoeba-style movement or interstitial migration, nally settle at the target site (Figure 1). The latter method relies mainly on cell surface receptors to receive molecules along a vascular extracellular signal concentration gradient, resulting in directional, non-blood-owdependent movement.15,91 Although the molecular mechanisms of stem cell homing are not yet completely clear, the phenomenon of stem cell homing in tissue repair and regeneration has received great attention from a number of scientists. It has become the core of in situ regeneration technology14,15,89 and has been successfully demonstrated in certain animal models.1630 The use of stem cell homing to achieve in situ tissue regeneration has unique advantages compared to current research on stem cell transplantation: (i) homing overcomes the limitations of stem cell transplantation and is benecial to clinical transformation;1315 (ii) stem cell homing induction factors and the GFs that promote stem cell proliferation and differentiation or other factors that play key roles in organizational integration can also be combined with tissueengineering-based porous scaffold materials86 to achieve an orderly, coordinated release and can be developed into commercial products for clinical use. The molecular and physical information coded within extracellular matrices is guiding the development of advanced biomimetic scaffolds for robust stem cell homing and tissue regeneration. Several powerful extracellular inuences that have been biologically identied have already found their way into cell-instructive scaffolds, while many others remain largely unexploited and are in need of further investigation.90Further synergy between cell biological and biomaterial technologies promises to enable the development of new generations of therapeutic biomaterials that can recruit endogenous stem/progenitor cells and subsequently guide their differentiation to form new tissues.92 Thus, bioscaffold materials with cell recruitment properties have broad application and translational potential. This is not only the focus and key of tissue engineering

12
Table 4. Potential Cell Homing Factors for Periodontal Tissue and Bone Regeneration Homing Factors SDF-1 bFGF BMP-2 VEGF PDGF-BB Target Cells (Determined thus Far)

Biotechnol. Prog., 2012, Vol. 28, No. 1

Ref. (s) 16,24,30 17,100,101 21,29,30 106 80,84

A wide range of cell types including stem/progenitor cells derived from bone marrow and circulating system Cells derived from periodontal ligament and dental pulp Bone precursor cells including bone marrow-derived cells Endothelial cells Smooth-muscle cells and pericycles

bFGF, basic broblast growth factor; BMP, bone morphogenetic protein; PDGF, platelet-derived growth factor; SDF-1, stromal cell-derived factor-1; VEGF, vascular endothelial growth factor.

research but also highlights essential scientic issues that must be addressed in endogenous regenerative medicine.91 Selection and combination of different factors The factors required for the regeneration of different tissues can vary. Guidance on which factors to deliver and the timing of delivery is emerging from advances in understanding of critical pathways involved in the development of integrated periodontal tissues. The selection and combination of factors are the rst important issues to be considered for the in vitro design of biomaterials for in situ periodontal tissue regeneration (Table 4). In terms of instructing stem cell homing, SDF-1 probably requires primary consideration. SDF-1 is a cytokine that belongs to the CXC class chemokine protein, with the system named CXCL12 (CXC chemokine ligand-12). It is the only known natural chemokine in the body that is activated by binding to CXCR4 receptors.93,94 Recent studies have shown that the SDF-1/CXCR4 complex plays a key role in stem cell homing. Under normal circumstances, SDF-1 is primarily released from bone marrow stromal cells to ensure that the CD34 HSCs are chemotactically adhered to the bone marrow. Results have shown that intravenous injection of SDF-1a can promote the mobilization of HSCs and that the neutralizing antibody antiCXCR4 and SDF-1 can inhibit chemotaxis.9497 HSCs, endothelial progenitor cells, MSCs and the like are capable of expressing the specic receptor CXCR4. The increase in local SDF-1 in damaged tissues is a key factor that regulates stem cell homing and recruitment to injured tissues. Therefore, SDF-1 can serve as an important induction factor for stem cell homing; its physiological function and effect have also been conrmed by many experiments.16,24,29,30,9397 bFGF is a heparin-binding mitogenic protein that can induce mesenchymal cell proliferation and differentiation. It is an important factor in promoting bone and periodontal tissue regeneration,98 which has been conrmed in clinical trials at multiple centers.99 Recent studies have also shown that bFGF stimulates the directed migration of PDL cells via PI3K/AKT signaling and CD44/hyaluronan interaction.100 More recently, bFGF was found to induce recellularization and re-vascularization in endodontically treated human teeth implanted into the dorsum of rats, suggesting that endogenous dental pulp cells, including stem/progenitor cells, may be recruited and subsequently differentiated by the chemotaxis of selective cytokines in the regeneration of dental pulp.101 BMP belongs to the TGF-b superfamily. As an osteogenic GF, BMP-2 is known to strongly stimulate bone formation, mainly by inducing recruitment to bone precursor cells21,22,29,102 and by promoting the angiogenesis and differentiation of MSCs. Results have demonstrated that BMP-2 can achieve complete bone regeneration in bone defect

regions and has a strong ability for ectopic bone induction.29,30,102 Thus, in the process of new bone formation, BMP-2 is both the inductor of cell recruitment and the stimulating factor that promotes cell differentiation. BMP-7, also known as osteogenic protein (OP)-1, is highly efcient at promoting osteoinductive activity. It can promote the regenerative repair of defects and the creation of hard tissue such as bone, cartilage, dentin, etc. At the same time, it can induce undifferentiated MSCs into bone cells, cartilage cells and cementum.103,104 In the process of periodontal tissue regeneration, BMP-2 and BMP-7 can play a synergistic role; the former signicantly promotes the formation of new bone, while the latter can induce the regeneration of cementum.105 In the process of angiogenesis, different GFs play important roles at different stages. Although vascular endothelial growth factor (VEGF) is the initiating factor for early angiogenesis, VEGF alone cannot form a mature vascular system.91 In contrast, PDGF-BB can recruit smooth muscle cells to promote vascular maturity and stability.106 Results have demonstrated that the controlled release of VEGF and PDGF-BB can effectively promote neovascularization (Figure 6).80,84 Analysis of data published to date indicates that SDF-1 is the most widely used homing inducer. The combined application of SDF-1 and BMP-2 has become the primary strategy used to induce MSC homing to promote bone regeneration;29 in the bone healing process, bFGF may stimulate the proliferation of early osteoblasts, and BMP-2 can enhance bone mineralization.107,108 Following periodontal wounding, rhBMP-2 may stimulate cell recruitment by increasing the proliferation and migration of cells from the adjacent unwounded PDL into the wounded area, thus promoting periodontal regeneration by stimulating the formation of new cementum.109 Studies have also shown that the orderly release of BMP-2 and BMP-7 may be coordinated in development and enhances new bone formation,83,105 and angiogenesis is the synergistic result of VEGF and PDGFBB. In the process of angiogenesis, bFGF and PDGF-BB can also play a synergistic role in promoting blood vessel maturation110 or in inducing vascular endothelial cells to differentiate into vascular forming cells.111 Given the various roles of key factors in periodontal tissue regeneration at different stages, the combination and screening of different factors are important areas of future research. By addressing the problem of cell recruitment design and the orderly release of multiple factors, we can apply periodontal tissue regeneration scaffold designs to the development of a new generation of biomaterials (Figure 7).

Sequential release of multiple factors Studies have shown that the administration of SDF-1 can signicantly extend the release time of drugs, thereby

Biotechnol. Prog., 2012, Vol. 28, No. 1

13

Figure 6. Schematic of the sequential release of vascular endothelial growth factor (VEGF) and platelet-derived growth factor BB (PDGF-BB) for vascularization (modied from Place et al.91).
New blood vessels initially form by endothelial cell organization into tubes with the help of VEGF. However, VEGF alone results in the formation of unstable, leaky vessels. The subsequent release of PDGF-BB helps to recruit smooth muscle cells and pericytes, which favor vessel maturation.

increasing the effect of cell recruitment and ensuring that they safely and effectively perform their biological functions.112115 After the local or systemic administration of SDF-1a, a problem arises involving its rapid diffusion and degradation by enzymes such as matrix metalloproteinase -2/ 9 or CD26/dipeptidyl peptidase IV (DPPIV). Hydrolyzed products such as nerve toxin SDF-1 (5-68) may be related to the pathogenesis of dementia.94 Meanwhile, cell-recruiting scaffold material must be designed with the ability to promote stem cell proliferation/differentiation and stimulate the formation of new blood vessels; only then can functional tissue regeneration occur.114,115 On the other hand, based on an analysis of the wound healing cascade, homing induction factors should function before proliferated and differentiated GFs, while vascular formation should occur during new tissue regeneration. Different factors should execute their functions during specic periods to then effectively increase the biological efcacy of biologically active drugs and reduce possible side effects. Unfortunately, no study has taken all of these factors into consideration thus far; there is also no research involving the remote activation of stem cells in the body to enhance local cell recruitment and thereby promote tissue regeneration (Figure 8). It is, of course, a substantial way to go before such a dream is to be realized, but there is room for hope. In recent years, the systemic administration of SDF-1 has been shown to mobilize and activate the bodys stem cell niche,9597 pointing to an important adjunctive strategy in the design of a future endogenous regeneration strategy that is worth deeper exploration.

Figure 7. Sequential release of multiple factors from hypothetical material devices designed for periodontal regeneration (medical hypotheses in need of further investigation).
Both the essential growth factors that determine the fate of multiple periodontal tissues and the criteria used to establish the doses demonstrate the molecular complexity of these devices. A few of the challenges facing the eld involve improving the pharmacokinetics and biodistribution of the growth factors released and developing tailored products for various pathological situations (SDF-1: stromal-derived factor-1; bFGF: basic broblast growth factors; BMP-2/7: bone morphogenetic protein-2/7; VEGF: vascular endothelial growth factor; PDGF-BB: platelet-derived growth factor BB).

Consideration of foreign body reaction Implanted biomedical devices, such as those based on polymeric materials, provoke the body to initiate an inammatory reaction to the surface of the implanted material. This is known as the foreign body reaction/response (FBR) and is potentially affected by the physical and chemical properties of implanted biomaterials; moreover, it has proven to be a hindrance to the functionality of biomaterials.116,117 Hence,

FBR must be carefully considered in the design of biomaterials for in situ periodontal regeneration. The events in the FBR include protein adsorption, monocyte/macrophage adhesion, macrophage fusion to form foreign body giant cells (FBGCs), consequences of the FBR on biomaterials and cross-talk between macrophages/FBGCs and inammatory/ wound healing cells. The FBR begins as wound healing, which includes the accumulation of exudate at the site of injury, inltration of inammatory cells to debride the area

14

Biotechnol. Prog., 2012, Vol. 28, No. 1

Figure 8. Schematic of the systemic delivery of directional cues (e.g., stromal cell-derived factor-1) that instruct stem cells from distant cell niches such as bone marrow to nd their way home (illustrations not to scale, modied from Chen et al.15).
Drug delivery system (DDS) composed of various natural and synthetic biocompatible polymers can provide controlled drug delivery by differing mechanisms. One technically simple DDS involved the encapsulation of cues into DDS (e.g., microparticulates), and the subsequent release of cues is programmed according to the design of the delivery vehicles (normally via cue diffusion and polymer degradation). After systemic administration of DDS, the sustained release of cues guides the mobilized stem cells journey through the blood and to the microvasculature at a target site (normally over distance), where the cells replenish and maintain the cell niche; hence they enhance the regenerative potential of the tissue or directly reach the site of injury and thereafter participate in tissue repair and regeneration.

and the production of particulates or other foreign bodies that inhibits full healing. The success of implanted medical devices is therefore dependent on the ability to direct this reaction via the material-dependent control of key cellular components, macrophages and FBGCs.118 Macrophages interact with the surfaces of implanted materials and secrete intercellular signals, including cytokines and GFs, which direct the actions of immune cells in the surrounding tissue. Understanding these complex inter-relationships is the key to designing a biomaterial that stimulates regeneration and induces tolerance in tissue engineering applications. It is now clear that the type and quantity of cytokines and GFs produced by macrophages at an implant surface could be indicators of the outcome of the FBR.119 Thus, it can be understood that macrophage adhesion to biomaterial surface is one of the early steps of the inammatory response to an implanted material, which indicates that a surface that modulates macrophage adhesion may serve to direct the FBR.120 Other variables that affect the host response include manufacturing processes, the rate of scaffold degradation and the presence of cross-species antigens.121 Although inammation is usually viewed as detrimental, it has unexpected and

potentially benecial effects on brosis and transplant rejection. For example, the magnitude of inammation due to a biomaterial does not determine the extent of brosis. Similarly, biomaterials do not always show adjuvancy.122 Although there remains much more to be done, the innate and adaptive immune responses of the host must be controlled to enable the positive function of the implanted biomaterials. The host response is also central to the remodeling that enables biomaterials to be integrated by the host. An in-depth understanding of how the immune system interacts with biomaterials and how biomaterials inuence these interactions may prove pivotal to the safety, biocompatibility and function of the device or system under consideration for any medical use.123 There is a long road ahead, but the benets warrant the effort. Importance of instructive design In addition to information transfer between different cell types, cells derive a vast wealth of information from their in vivo milieu, including the biomaterials that surround and separate them within tissues.91 The native ECM surrounded,

Biotechnol. Prog., 2012, Vol. 28, No. 1

15

and produced, by cells is instructive, providing a dynamic and spatially heterogeneous constellation of microstructural, compositional and mechanical cues that can inuence cell behavior by binding and clustering cell surface integrins, which act as receptors for triggering signal transduction pathways.42 Implantable biomaterials must take on this instructive role to some degree in order to mimic the natural molecular, mechanical and architectural components of the ECM, and hence, to maintain cell viability and control cell fate toward safe and effective tissue regeneration.91 The ECM provides an important model for biomaterials design. Most ECM components such as collagen, elastin, laminin, bronectin, glycoproteins and proteoglycans have small bioactive domains for cell binding, proteolytic degradation and/or GF-binding. Usually, from these domains, short peptide sequences can be identied as motifs that are responsible for these biofunctions.124 To this end, bioactive biomaterials have been achieved typically by incorporating short peptide sequences into suitable materials to emulate key molecular features of the ECM either by surface modication or by bulk incorporation, and clues for their design come from naturally bioactive scaffolds such as collagen and brin.125 Unlike the entire protein structure, which is subject to denaturation and degradation, short peptide sequences have the advantage of being relatively stable for modication, tunable for cell binding and easy to be synthesized in a large scale. The modication of biomaterials with adhesive peptides can provide biological ligands for cell-scaffold interactions that promote cell adhesion, proliferation, differentiation, osteogenesis and osseointegration, wherein the integrin-binding arginine-glycine-aspartic acid (RGD) sequence found in many ECM proteins, including bronectin, laminin, collagen type IV, tenascin and thrombospondin is by far the most effective and most often employed peptide sequence for stimulated cell adhesion on synthetic surfaces.126 This is based upon its widespread distribution and use throughout the organism, its ability to address more than one cell adhesion receptor and its biological impact on cell anchoring, behavior and survival.126 Substantial evidence suggests that RGD modication is sufcient to transform articial construct from a relatively inert biomaterial into a substance that supports the formation of convincing tissuelike structures in vitro and in vivo.127130 The effects of RGD, however, can depend on the specic application and might diminish during its inevitable interaction with blood and tissue proteins upon implantation. In terms of periodontal and bone regeneration, other adhesive factors such as heparin-sulfate-mediated factors, consensus-heparin-binding motifs, and arginineglycine-aspartate-serine may be more specic for osteoblast adhesion than RGD.131,132 Besides, the degradation rate of biomaterials represents one of most important considerations and once implanted, it is highly desirable if the degradation rate matches with new tissue regeneration at site of injury.124 Keeping this in mind, synthetic biomaterials have been crosslinked by enzymedegradable peptide sequences to mimic the invasive characteristics of a native provisional ECM, where a combination of integrin-binding sites and substrates for matrix metalloproteinases (MMP) was required to render the networks degradable and invasive by cells via cell-secreted MMPs, allowing the cells to migrate through the scaffold in a process reminiscent of tissue remodeling.133 Cell-mediated degradation is more likely to generate a biomaterial temporal prole in tune with the generation of new tissue. These bio-

materials may be useful in tissue engineering and cell biology as alternatives for naturally occurring ECM-derived materials such as brin or collagen.133 Bioactive peptides for molecular engineering of instructive biomaterials can be obtained from biological or chemically synthesized sources, increasing their specic cellular responses for tissue growth and development. Compared to using an entire protein in regenerative therapy, these peptides demonstrate potential advantages such as overcoming possible immunogenicity, being less susceptible to degradation and producing fewer tumor-related side effects.124 The promise of biosynthetic approaches to biomaterials design, however, must be weighed against the fact that very little is known about the in vivo performance of systems prepared in this way. Although the chemical composition has been the focus of biomaterials design for the past few decades, there is growing appreciation of the importance of other properties (e.g. topological, mechanical and electrical cues) that are important in instructing a biological response effectively for a given tissue; each tissue has its own characteristic mechanical properties that vary over the microscale and inuence cellular behavior.134 The cellular response in vitro and in vivo can drastically alter depending on the mechanical properties of biomaterials. On a basic level, the scaffold must usually provide some level of physical support from the moment of implantation, to assist tissue function while new matrix is being deposited.91 Ideally, the scaffold should have mechanical properties consistent with the anatomical site into which it is to be implanted and, from a practical perspective, it must be strong enough to allow surgical handling during implantation. Key roles in molecular pathways are played by adhesion complexes and the actin-myosin cytoskeleton, whose contractile forces are transmitted through transcellular structures.135 Therefore, the mechanical properties of the substrate to which the cells are attached are critical to the regulation of cellular mechanotransduction and subsequent cellular behavior.136 Advances in biomaterials will include extensive development of more sophisticated medical devices with respect to biofunctionality and the expanded application of developed biomaterials into new biomedical elds such as Periodontology. In future, understanding the way in which complex dynamic behaviors are accomplished in nature and rethinking fundamentally the way in which inspiration is drawn from biology may lead to the design of novel biomaterials that mimic nature not through presenting active motifs replicated exactly from biological molecules but rather through reproducing the functional behavior of these biomaterials to obtain properties that are currently unavailable.134,137

Future perspectives The development of functional biomaterials/scaffolds for cell delivery is a critical research focus of tissue engineering and regenerative medicine.45,84,136,138141 However, to date, the majority of cell-based therapies have not yet been applied to patients. Most studies remain at the laboratory stage of research, and their translation to clinical practice faces severe difculties and challenges.37 Mao and coworkers have recently developed a new path using biofunctionalized biomaterials to instruct host stem cell homing to the site of injury,1618 which provides a classic example of

16

Biotechnol. Prog., 2012, Vol. 28, No. 1

the recruitment of a patients own cells for endogenous tissue regeneration. This strategy promises major improvements and the potential to be adapted to the regeneration of pulp and periodontal tissue.1316,142 Not only is there the controversial prospect of the clinical application of current stem cell therapy and tissue engineering technology in the treatment of non-fatal diseases, but there is also a concern over whether high-risk technology that requires signicant investment should be applied to treat a disease that can be treated with current alternative methods (such as tooth extraction and dental implants).14 The emergence and development of cell recruitment regeneration technology will surely bring new vitality to regenerative dentistry. Although the mechanisms of current biological signaling molecules and functional biological materials that promote stem cell homing require further study, the results of previous studies indicate that this line of research has certainly become an important strategy for endogenous regenerative medicine.15 The research of Mao and coworkers only combined stem cell homing induction factors with biomaterial scaffolds directly, and the results were only obtained for small animals.1518 Improvements in the induction of directional migration, recruitment, settlement, differentiation and molding of the patients own functional cells are still required. Indeed, the formation of new functional tissues in large animals and even in human patients has yet to be demonstrated.15 In further studies in endogenous periodontal regeneration, the optimal combination of factors and dose criteria and the design of cellular scaffolds must be investigated; additionally, these studies should take into account the physical and chemical properties and structural features of the scaffold to achieve the sequential controlled release of multiple factors (Figure 9).86 All of these issues must be concisely integrated into research efforts in the future, with a focus on circumventing each issue one after another.143,144 Ultimately, clinicians will have commercialized products to aid patients with periodontal disease.

Figure 9. Implantable biomaterials developed in our laboratory for the management of periodontal defects (unpublished data).
(A) An image of a biofunctional scaffold fabricated from glycidyl methacrylate derivatized dextran (Dex-GMA) and gelatin (Gtn), wherein Dex-GMA/Gtn microparticles loaded with growth factors are incorporated into its macroporous pore structures; (B) cutaway view of the highly porous interconnected structures and the microencapsulated growth factors entrapped within the inner pores of the hybrid scaffold.

logical organs, which has important clinical signicance and wide application potential.

Conclusions
In summary, the design of biomaterials for cell recruitment, the selection and combination of multiple factors with biomaterials and their dosages and controlled release criteria are important foundations of the new eld of functionalized scaffold research in periodontal regenerative medicine. The use of drug delivery methods to enable the coordinated and orderly controlled release of stem cell homing inducers, stem cell proliferation and differentiation factors and new blood vessel formation stimulators from next generation of medical devices are expected to allow robust mobilization and homing of resident stem cells to periodontal defect areas, where the coordinated interactions with various signaling molecules, other cells and their articial microenvironments may dene a local biochemical and mechanical niche with complex and dynamic regulation that leads to safe and effective regeneration of multiple tooth-supporting tissues. Research on the design of new biofunctionalized scaffolds will provide an effective basis for the study of endogenous regeneration of various tissues and in vivo tissue engineering. Furthermore, it will provide new treatments for the reconstruction of complex tissues and the structure and function of physio-

Acknowledgment
Fundings from the National Natural Science Foundation of China (81071253/H1818; 31170912/C1002) for some of the work mentioned in the review is gratefully appreciated.

Literature Cited
1. Pihlstrom BL, Michalowicz BS, Johnson NW. Periodontal diseases. Lancet. 2005;366:18091820. UM. Periodontal 2. Lee J, Stavropoulos A, Susin C, Wikesjo regeneration: focus on growth and differentiation factors. Dent Clin North Am. 2010;54:93111. 3. Seo BM, Miura M, Gronthos S, Bartold PM, Batouli S, Brahim J, Young M, Robey PG, Wang CY, Shi S. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet. 2004;364:149155. 4. Liu Y, Zheng Y, Ding G, Fang D, Zhang C, Bartold PM, Gronthos S, Shi S, Wang S. Periodontal ligament stem cellmediated treatment for periodontitis in miniature swine. Stem Cells. 2008;26:10651073. 5. Ding G, Liu Y, Wang W, Wei F, Liu D, Fan Z, An Y, Zhang C, Wang S. Allogeneic periodontal ligament stem cell therapy for periodontitis in swine. Stem Cells. 2010;28:18291838.

Biotechnol. Prog., 2012, Vol. 28, No. 1


6. Tsumanuma Y, Iwata T, Washio K, Yoshida T, Yamada A, Takagi R, Ohno T, Lin K, Yamato M, Ishikawa I, Okano T, Izumi Y. Comparison of different tissue-derived stem cell sheets for periodontal regeneration in a canine 1-wall defect model. Biomaterials. 2011;32:58195825. 7. Kawaguchi H, Hirachi A, Hasegawa N, Iwata T, Hamaguchi H, Shiba H, Takata T, Kato Y, Kurihara H. Enhancement of periodontal tissue regeneration by transplantation of bone marrow mesenchymal stem cells. J Periodontol. 2004;75:12811287. 8. Hasegawa N, Kawaguchi H, Hirachi A, Takeda K, Mizuno N, Nishimura M, Koike C, Tsuji K, Iba H, Kato Y, Kurihara H. Behavior of transplanted bone marrow-derived mesenchymal stem cells in periodontal defects. J Periodontol. 2006;77:1003 1007. 9. Li H, Yan F, Lei L, Li Y, Xiao Y. Application of autologous cryopreserved bone marrow mesenchymal stem cells for periodontal regeneration in dogs. Cells Tissues Organs. 2009;190: 94101. 10. Yang Y, Rossi FM, Putnins EE. Periodontal regeneration using engineered bone marrow mesenchymal stromal cells. Biomaterials. 2010;31:85748582. 11. Tobita M, Uysal AC, Ogawa R, Hyakusoku H, Mizuno H. Periodontal tissue regeneration with adipose-derived stem cells. Tissue Eng Part A. 2008;14:945953. 12. Duan X, Tu Q, Zhang J, Ye J, Sommer C, Mostoslavsky G, Kaplan D, Yang P, Chen J. Application of induced pluripotent stem (iPS) cells in periodontal tissue regeneration. J Cell Physiol. 2011;226:150157. 13. Mao JJ, Stosich MS, Moioli EK, Lee CH, Fu SY, Bastian B, Eisig SB, Zemnick C, Ascherman J, Wu J, Rohde C, Ahn J. Facial reconstruction by biosurgery: cell transplantation versus cell homing. Tissue Eng Part B Rev. 2010;16:257262. 14. Chen FM, Zhang J, Zhang M, An Y, Chen F, Wu ZF. A review on endogenous regenerative technology in periodontal regenerative medicine. Biomaterials. 2010;31:78927927. 15. Chen FM, Wu LA, Zhang M, Zhang R, Sun HH. Homing of endogenous stem/pregenitor cells for in situ tissue regeneration: promises, strategies, and translational perspectives. Biomaterials. 2011;32:31893209. 16. Kim K, Lee CH, Kim BK, Mao JJ. Anatomically shaped tooth and periodontal regeneration by cell homing. J Dent Res. 2010;89:842847. 17. Kim J, Xin X, Moioli EK, Chung J, Lee CH, Chen M, Fu SY, Koch PD, Mao J. Regeneration of dental pulp-like tissue by chemotaxis-induced cell homing. Tissue Eng Part A. 2010;16: 30233031. 18. Lee CH, Cook JL, Mendelson A, Moioli EK, Yao H, Mao JJ. Regeneration of the articular surface of the rabbit synovial joint by cell homing: a proof of concept study. Lancet. 2010; 376:440448. 19. Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, Capla JM, Galiano RD, Levine JP, Gurtner GC. Progenitor cell trafcking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10:858864. 20. Jin DK, Shido K, Kopp HG, Petit I, Shmelkov SV, Young LM, Hooper AT, Amano H, Avecilla ST, Heissig B, Hattori K, Zhang F, Hicklin DJ, Wu Y, Zhu Z, Dunn A, Salari H, Werb Z, Hackett NR, Crystal RG, Lyden D, Rai S. Cytokine-mediated deployment of SDF-1 induces revascularization through recruitment of CXCR4 hemangiocytes. Nat Med. 2006;12:557667. 21. Otsuru S, Tamai K, Yamazaki T, Yoshikawa H, Kaneda Y. Bone marrow-derived osteoblast progenitor cells in circulating blood contribute to ectopic bone formation in mice. Biochem Biophys Res Commun. 2007;354:453458. 22. Otsuru S, Tamai K, Yamazaki T, Yoshikawa H, Kaneda YA. Circulating bone marrow-derived osteoblast progenitor cells are recruited to the bone-forming site by the CXCR4/stromal cell-derived factor-1 pathway. Stem Cells. 2008;26:223234. 23. Kumagai K, Vasanji A, Drazba JA, Butler RS, Muschler GF. Circulating cells with osteogenic potential are physiologically mobilized into the fracture healing site in the parabiotic mice model. J Orthop Res. 2008;26:165175. 24. Thieme S, Ryser M, Gentsch M, Navratiel K, Brenner S, lng J, Gelinsky M, Ro sen-Wolff A. Stromal Stiehler M, Ro

17
cell-derived factor-1alpha-directed chemoattraction of transiently CXCR4-overexpressing bone marrow stromal cells into functionalized three-dimensional biomimetic scaffolds. Tissue Eng Part C Methods. 2009;15:687696. Lee DY, Cho TJ, Lee HR, Park MS, Yoo WJ, Chung CY, Choi IH. Distraction osteogenesis induces endothelial progenitor cell mobilization without inammatory response in man. Bone. 2010;46:673679. Shen W, Chen X, Chen J, Yin Z, Heng BC, Chen W, Ouyang HW. The effect of incorporation of exogenous stromal cell-derived factor-1 alpha within a knitted silk-collagen sponge scaffold on tendon regeneration. Biomaterials. 2010;31:72397249. rfer KI, Walter I, Kleiter M, Sandgren EP, Erben RG. Odo Role of endogenous bone marrow cells in long-term repair mechanisms after myocardial infarction. J Cell Mol Med. 2008;12:28672874. Zaruba MM, Theiss HD, Vallaster M, Mehl U, Brunner S, David R, Fischer R, Krieg L, Hirsch E, Huber B, Nathan P, Israel L, Imhof A, Herbach N, Assmann G, Wanke R, Mueller-Hoecker J, Steinbeck G, Franz WM. Synergy between CD26/DPP-IV inhibition and G-CSF improves cardiac function after acute myocardial infarction. Cell Stem Cell. 2009;4:313323. Kimura Y, Miyazaki N, Hayashi N, Otsuru S, Tamai K, Kaneda Y, Tabata Y. Controlled release of bone morphogenetic protein-2 enhances recruitment of osteogenic progenitor cells for de novo generation of bone tissue. Tissue Eng Part A. 2010;16:12631270. Ratanavaraporn J, Furuya H, Kohara H, Tabata Y. Synergistic effects of the dual release of stromal cell-derived factor-1 and bone morphogenetic protein-2 from hydrogels on bone regeneration. Biomaterials. 2011;32:27972811. Pizzo G, Guiglia R, Lo Russo L, Campisi G. Dentistry and internal medicine: from the focal infection theory to the periodontal medicine concept. Eur J Intern Med. 2010;21:496502. Al-Shammari KF, Al-Khabbaz AK, Al-Ansari JM, Neiva R, Wang HL. Risk indicators for tooth loss due to periodontal disease. J Periodontol. 2005;76:19101918. Muzzi L, Nieri M, Cattabriga M, Rotundo R, Cairo F, Pini Prato GP. The potential prognostic value of some periodontal factors for tooth loss: a retrospective multilevel analysis on periodontal patients treated and maintained over 10 years. J Periodontol. 2006;77:20842089. Chen FM, Jin Y. Periodontal tissue engineering and regeneration: current approaches and expanding opportunities. Tissue Eng Part B Rev. 2010;16:219255. Cao CF. Translational medicine: a new concept to guide the prevention and treatment of periodontal diseases. Zhonghua Kou Qiang Yi Xue Za Zhi. 2009;44:321323. Villar CC, Cochran DL. Regeneration of periodontal tissues: guided tissue regeneration. Dent Clin North Am. 2010;54:73 92. Reynolds MA, Aichelmann-Reidy ME, Branch-Mays GL. Regeneration of periodontal tissue: bone replacement grafts. Dent Clin North Am. 2010;54:5571. Rayment EA, Williams DJ. Concise review: mind the gap: challenges in characterizing and quantifying cell- and tissue-based therapies for clinical translation. Stem Cells. 2010;28: 9961004. Discher DE, Mooney DJ, Zandstra PW. Growth factors, matrices, and forces combine and control stem cells. Science. 2009; 324:16731677. Chen FM, Shelton RM, Jin Y, Chapple IL. Localized delivery of growth factors for periodontal tissue regeneration: role, strategies, and perspectives. Med Res Rev. 2009;29:472513. Kaigler D, Cirelli JA, Giannobile WV. Growth factor delivery for oral and periodontal tissue engineering. Expert Opin Drug Deliv. 2006;3:647662. Tayalia P, Mooney DJ. Controlled growth factor delivery for tissue engineering. Adv Mater. 2009;21:32693285. nchez M, Orive G. Potential of endogenous regenAnitua E, Sa erative technology for in situ regenerative medicine. Adv Drug Delivery Rev. 2010;62:741752. Vasita R, Katti DS. Growth factor-delivery systems for tissue engineering: a materials perspective. Expert Rev Med Devices. 2006;3:2947.

25.

26.

27.

28.

29.

30.

31. 32.

33.

34. 35. 36. 37. 38.

39. 40. 41.

42. 43. 44.

18
45. Andreadis ST, Geer DJ. Biomimetic approaches to protein and gene delivery for tissue regeneration. Trends Biotechnol. 2006; 24:331337. 46. Tabata Y. Signicance of release technology in tissue engineering. Drug Discov Today. 2005;10:16391646. 47. Guldberg RE. Spatiotemporal delivery strategies for promoting musculoskeletal tissue regeneration. J Bone Miner Res. 2009; 24:15071511. nchez M, Orive G, Andia I. Delivering growth fac48. Anitua E, Sa tors for therapeutics. Trends Pharmacol Sci. 2008;29:3741. 49. Kolambkar YM, Dupont KM, Boerckel JD, Huebsch N, Mooney DJ, Hutmacher DW, Guldberg RE. An alginate-based hybrid system for growth factor delivery in the functional repair of large bone defects. Biomaterials. 2011;32:6574. 50. Yan XZ, Ge SH, Sun QF, Guo HM, Yang PS. A pilot study evaluating the effect of recombinant human bone morphogenetic protein-2 and recombinant human beta-nerve growth factor on the healing of Class III furcation defects in dogs. J Periodontol. 2010;81:12891298. 51. Park YJ, Lee YM, Park SN, Sheen SY, Chung CP, Lee SJ. Platelet derived growth factor releasing chitosan sponge for periodontal bone regeneration. Biomaterials. 2000;21:153159. 52. Sigurdsson TJ, Nygaard L, Tatakis DN, Fu E, Turek TJ, Jin L, UM. Periodontal repair in dogs: evaluaWozney JM, Wikesjo tion of rhBMP-2 carriers. Int J Periodontics Restorative Dent. 1996;16:524537. 53. Giannobile WV, Hernandez RA, Finkelman RD, Ryan S, Kiritsy CP, DAndrea M, Lynch SE. Comparative effects of platelet-derived growth factor-BB and insulin-like growth factor-I, individually and in combination, on periodontal regeneration in Macaca fascicularis. J Periodontal Res. 1996; 31:301312. 54. Kinoshita A, Oda S, Takahashi K, Yokota S, Ishikawa I. Periodontal regeneration by application of recombinant human bone morphogenetic protein-2 to horizontal circumferential defects created by experimental periodontitis in beagle dogs. J Periodontol. 1997;68:103109. 55. Blumenthal NM, Koh-Kunst G, Alves ME, Miranda D, Soren UM. Effect of surgical implansen RG, Wozney JM, Wikesjo tation of recombinant human bone morphogenetic protein-2 in a bioabsorbable collagen sponge or calcium phosphate putty carrier in intrabony periodontal defects in the baboon. J Periodontol. 2002;73:14941506. 56. Herberg S, Siedler M, Pippig S, Schuetz A, Dony C, Kim CK, UM. Development of an injectable composite as a Wikesjo carrier for growth factor-enhanced periodontal regeneration. J Clin Periodontol. 2008;35:976984. UM, Park JC, Chae GJ, Pippig SD, Bastone 57. Min CK, Wikesjo P, Kim CS, Kim CK. Wound healing/regeneration using recombinant human growth/differentiation factor-5 in an injectable poly-lactide-co-glycolide-acid composite carrier and a one-wall intra-bony defect model in dogs. J Clin Periodontol. 2011;38:261268. 58. Annabi N, Nichol JW, Zhong X, Ji C, Koshy S, Khademhosseini A, Dehghani F. Controlling the porosity and microarchitecture of hydrogels for tissue engineering. Tissue Eng Part B Rev. 2010;16:371383. 59. Chen FM, Zhao YM, Sun HH, Jin T, Wang QT, Zhou W, Wu ZF, Jin Y. Novel glycidyl methacrylated dextran (Dex-GMA)/ gelatin hydrogel scaffolds containing microspheres loaded with bone morphogenetic proteins: formulation and characteristics. J Control Release. 2007;118:6577. 60. Choi DH, Park CH, Kim IH, Chun HJ, Park K, Han DK. Fabrication of core-shell microcapsules using PLGA and alginate for dual growth factor delivery system. J Control Release. 2010;147:193201. 61. Chen FM, Zhao YM, Zhang R, Jin T, Sun HH, Wu ZF, Jin Y. Periodontal regeneration using novel glycidyl methacrylated dextran (Dex-GMA)/gelatin scaffolds containing microspheres loaded with bone morphogenetic proteins. J Control Release. 2007;121:8190. UM, Qahash M, Thomson RC, Cook AD, Rohrer 62. Wikesjo MD, Wozney JM, Hardwick WR. Space-providing expanded polytetrauoroethylene devices dene alveolar augmentation at

Biotechnol. Prog., 2012, Vol. 28, No. 1


dental implants induced by recombinant human bone morphogenetic protein 2 in an absorbable collagen sponge carrier. Clin Implant Dent Relat Res. 2003;5:112123. Chen FM, Chen R, Wang XJ, Sun HH, Wu ZF. In vitro cellular responses to scaffolds containing two microencapulated growth factors. Biomaterials. 2009;30:52155224. Elangovan S, Karimbux N. Review paper: DNA delivery strategies to promote periodontal regeneration. J Biomater Appl. 2010;25:318. Zhang Y, Shi B, Li C, Wang Y, Chen Y, Zhang W, Luo T, Cheng X. The synergetic bone-forming effects of combinations of growth factors expressed by adenovirus vectors on chitosan/collagen scaffolds. J Control Release. 2009;136:172178. Chang PC, Seol YJ, Cirelli JA, Pellegrini G, Jin Q, Franco LM, Goldstein SA, Chandler LA, Sosnowski B, Giannobile WV. PDGF-B gene therapy accelerates bone engineering and oral implant osseointegration. Gene Ther. 2010;17:95104. Chen FM, Ma ZW, Wang QT, Wu ZF. Gene delivery for periodontal tissue Engineering: current knowledgefuture possibilities. Curr Gene Ther. 2009;9:248266. Chang PC, Cirelli JA, Jin Q, Seol YJ, Sugai JV, DSilva NJ, Danciu TE, Chandler LA, Sosnowski BA, Giannobile WV. Adenovirus encoding human platelet-derived growth factor-B delivered to alveolar bone defects exhibits safety and biodistribution proles favorable for clinical use. Hum Gene Ther. 2009;20:486496. Lee SH, Shin H. Matrices and scaffolds for delivery of bioactive molecules in bone and cartilage tissue engineering. Adv Drug Delivery Rev. 2007;59:339359. Uebersax L, Merkle HP, Meinel L. Biopolymer-based growth factor delivery for tissue repair: from natural concepts to engineered systems. Tissue Eng Part B Rev. 2009;15:263289. Biondi M, Ungaro F, Quaglia F, Netti PA. Controlled drug delivery in tissue engineering. Adv Drug Delivery Rev. 2008; 60:229242. Nagaoka M, Jiang HL, Hoshiba T, Akaike T, Cho CS. Application of recombinant fusion proteins for tissue engineering. Ann Biomed Eng. 2010;38:683693. Voog J, Jones DL. Stem cells and the niche: a dynamic duo. Cell Stem Cell. 2010;6:103115. Schroeder T. Imaging stem-cell-driven regeneration in mammals. Nature. 2008;453:345351. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453:314321. Lee K, Silva EA, Mooney DJ. Growth factor delivery-based tissue engineering: general approaches and a review of recent developments. J R Soc Interface. 2011;8:153170. Moioli EK, Clark PA, Xin X, Lal S, Mao JJ. Matrices and scaffolds for drug delivery in dental, oral and craniofacial tissue engineering. Adv Drug Delivery Rev. 2007; 59:308324. Marklein RA, Burdick JA. Controlling stem cell fate with material design. Adv Mater. 2010;22:175189. Domachuk P, Tsioris K, Omenetto FG, Kaplan DL. Bio-microuidics: biomaterials and biomimetic designs. Adv Mater. 2010; 22:249260. Richardson TP, Peters MC, Ennett AB, Mooney DJ. Polymeric system for dual growth factor delivery. Nat Biotechnol. 2001; 19:10291034. Borselli C, Storrie H, Benesch-Lee F, Shvartsman D, Cezar C, Lichtman JW, Vandenburgh HH, Mooney DJ. Functional muscle regeneration with combined delivery of angiogenesis and myogenesis factors. Proc Natl Acad Sci USA. 2010;107: 32873292. Kanczler JM, Ginty PJ, White L, Clarke NM, Howdle SM, Shakesheff KM, Oreffo RO. The effect of the delivery of vascular endothelial growth factor and bone morphogenic protein-2 to osteoprogenitor cell populations on bone formation. Biomaterials. 2010;31:12421250. Yilgor P, Hasirci N, Hasirci V. Sequential BMP-2/BMP-7 delivery from polyester nanocapsules. J Biomed Mater Res A. 2010;93:528536. Chen RR, Silva EA, Yuen WW, Mooney DJ. Spatio-temporal VEGF and PDGF delivery patterns blood vessel formation and maturation. Pharm Res. 2007;24:258264.

63. 64. 65.

66.

67. 68.

69. 70. 71. 72. 73. 74. 75. 76. 77.

78. 79. 80. 81.

82.

83. 84.

Biotechnol. Prog., 2012, Vol. 28, No. 1


85. Wang X, Wenk E, Zhang X, Meinel L, Vunjak-Novakovic G, Kaplan DL. Growth factor gradients via microsphere delivery in biopolymer scaffolds for osteochondral tissue engineering. J Control Release. 2009;134:8190. 86. Chen FM, Zhang M, Wu ZF. Toward delivery of multiple growth factors in tissue engineering. Biomaterials. 2010;31:6279 6308. 87. Chen W, Tabata Y, Tong YW. Fabricating tissue engineering scaffolds for simultaneous cell growth and drug delivery. Curr Pharm Des. 2010;16:23882394. 88. Huang S, Fu X. Naturally derived materials-based cell and drug delivery systems in skin regeneration. J Control Release. 2010;142:149159. 89. Chen FM, An Y, Zhang R, Zhang M. New insights into and novel applications of release technology in periodontal reconstructive therapies. J Control Release. 2011;149:92110. 90. Laird DJ, von Andrian UH, Wagers AJ. Stem cell trafcking in tissue development, growth, and disease. Cell. 2008;132: 612630. 91. Place ES, Evans ND, Stevens MM. Complexity in biomaterials for tissue engineering. Nat Mater. 2009;8:457470. 92. Lutolf MP, Gilbert PM, Blau HM. Designing materials to direct stem-cell fate. Nature. 2009;462:433441. 93. Lau TT, Wang DA. Stromal cell-derived factor-1 (SDF-1): homing factor for engineered regenerative medicine. Expert Opin Biol Ther. 2011;11:189197. hlstedt S, Ozcelik C, Bader M. SDF-1a as a 94. Ghadge S, Mu therapeutic stem cell homing factor in myocardial infarction. Pharmacol Ther. 2011;129:97108. 95. Carr AN, Howard BW, Yang HT, Eby-Wilkens E, Loos P, Varbanov A, Qu A, DeMuth JP, Davis MG, Proia A, Terjung RL, Peters KG. Efcacy of systemic administration of SDF-1 in a model of vascular insufciency: support for an endothelium-dependent mechanism. Cardiovasc Res. 2006;69:925935. bel R, Li W, Pittermann E, Ugurlu96. Klopsch C, Furlani D, Ga can M, Kundt G, Zingler C, Titze U, Wang W, Ong LL, Wagner K, Li RK, Ma N, Steinhoff G. Intracardiac injection of erythropoietin induces stem cell recruitment and improves cardiac functions in a rat myocardial infarction model. J Cell Mol Med. 2009;13:664679. 97. erfoot SM, Andonegui G, Bonder CS, Liu L. Exogenous stromal cell-derived factor-1 induces modest leukocyte recruitment in vivo. Am J Physiol Heart Circ Physiol. 2008; 294:H2524H2534. 98. Qu D, Li J, Li Y, Gao Y, Zuo Y, Hsu Y, Hu J. Angiogenesis and osteogenesis enhanced by bFGF ex vivo gene therapy for bone tissue engineering in reconstruction of calvarial defects. J Biomed Mater Res A. 2011;96:543551. 99. Kitamura M, Akamatsu M, Machigashira M, Hara Y, Sakagami R, Hirofuji T, Hamachi T, Maeda K, Yokota M, Kido J, Nagata T, Kurihara H, Takashiba S, Sibutani T, Fukuda M, Noguchi T, Yamazaki K, Yoshie H, Ioroi K, Arai T, Nakagawa T, Ito K, Oda S, Izumi Y, Ogata Y, Yamada S, Shimauchi H, Kunimatsu K, Kawanami M, Fujii T, Furuichi Y, Furuuchi T, Sasano T, Imai E, Omae M, Yamada S, Watanuki M, Murakami S. FGF-2 stimulates periodontal regeneration: results of a multi-center randomized clinical trial. J Dent Res. 2011;90:3540. 100. Shimabukuro Y, Terashima H, Takedachi M, Maeda K, Nakamura T, Sawada K, Kobashi M, Awata T, Oohara H, Kawahara T, Iwayama T, Hashikawa T, Yanagita M, Yamada S, Murakami S. Fibroblast growth factor-2 stimulates directed migration of periodontal ligament cells via PI3K/AKT signaling and CD44/hyaluronan interaction. J Cell Physiol. 2011; 226:809821. 101. Suzuki T, Lee CH, Chen M, Zhao W, Fu SY, Qi JJ, Chotkowski G, Eisig SB, Wong A, Mao JJ. Induced migration of dental pulp stem cells for in vivo pulp regeneration. J Dent Res. 2011;90:10131018. 102. Fong EL, Chan CK, Goodman SB. Stem cell homing in musculoskeletal injury. Biomaterials. 2011;32:395409. 103. Zhang Y, Song J, Shi B, Wang Y, Chen X, Huang C, Yang X, Xu D, Cheng X, Chen X. Combination of scaffold and adenovirus vectors expressing bone morphogenetic protein-7 for al-

19
veolar bone regeneration at dental implant defects. Biomaterials. 2007;28:46354642. Hakki SS, Foster BL, Nagatomo KJ, Bozkurt SB, Hakki EE, Somerman MJ, Nohutcu RM. Bone morphogenetic protein-7 enhances cementoblast function, in vitro. J Periodontol. 2010; 81:16631674. Ripamonti U, Crooks J, Petit JC, Rueger DC. Periodontal tissue regeneration by combined applications of recombinant human osteogenic protein-1 and bone morphogenetic protein-2. A pilot study in Chacma baboons (Papio ursinus). Eur J Oral Sci. 2001:109:241248. Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specic growth factors and blood vessel formation. Nature. 2000;407:242248. Otsuki S, Hanson SR, Miyaki S, Grogan SP, Kinoshita M, Asahara H, Wong CH, Lotz MK. Extracellular sulfatases support cartilage homeostasis by regulating BMP and FGF signaling pathways. Proc Natl Acad Sci USA. 2010;107:10202 10207. Hughes-Fulford M, Li CF. The role of FGF-2 and BMP-2 in regulation of gene induction, cell proliferation and mineralization. J Orthop Surg Res. 2011;6:8. King GN, Hughes FJ. Bone morphogenetic protein-2 stimulates cell recruitment and cementogenesis during early wound healing. J Clin Periodontol. 2001;28:465475. kenhielm E, Pawliuk R, Wariaro D, Post MJ, WahlCao R, Bra berg E, Leboulch P, Cao Y. Angiogenic synergism, vascular stability and improvement of hind-limb ischemia by a combination of PDGF-BB and FGF-2. Nat Med. 2003;9:604613. De Marchis F, Ribatti D, Giampietri C, Lentini A, Faraone D, Scoccianti M, Capogrossi MC, Facchiano A. Platelet-derived growth factor inhibits basic broblast growth factor angiogenic properties in vitro and in vivo through its alpha receptor. Blood. 2002;99:20452053. Segers VF, Tokunou T, Higgins LJ, MacGillivray C, Gannon J, Lee RT. Local delivery of protease-resistant stromal cell derived factor-1 for stem cell recruitment after myocardial infarction. Circulation. 2007;116:16831692. Zhang G, Nakamura Y, Wang X, Hu Q, Suggs LJ, Zhang J. Controlled release of stromal cell-derived factor-1 alpha in situ increases c-kit cell homing to the infarcted heart. Tissue Eng. 2007;13:20632071. He X, Ma J, Jabbari E. Migration of marrow stromal cells in response to sustained release of stromal-derived factor-1alpha from poly(lactide ethylene oxide fumarate) hydrogels. Int J Pharm. 2010;390:107116. Kimura Y, Tabata Y. Controlled release of stromal cell-derived factor-1 from gelatin hydrogels enhances angiogenesis. J Biomater Sci Polym Ed. 2010;21:3751. Ye Q, Harmsen MC, van Luyn MJ, Bank RA. The relationship between collagen scaffold cross-linking agents and neutrophils in the foreign body reaction. Biomaterials. 2010;31:9192 9201. Ye Q, van Amerongen MJ, Sandham JA, Bank RA, van Luyn MJ, Harmsen MC. Site-specic tissue inhibitor of metalloproteinase-1 governs the matrix metalloproteinases-dependent degradation of crosslinked collagen scaffolds and is correlated with interleukin-10. J Tissue Eng Regen Med. 2011;5:264 274. Khandwekar AP, Patil DP, Hardikar AA, Shouche YS, Doble M. In vivo modulation of foreign body response on polyurethane by surface entrapment technique. J Biomed Mater Res A. 2010;95:413423. Mesure L, De Visscher G, Vranken I, Lebacq A, Flameng W. Gene expression study of monocytes/macrophages during early foreign body reaction and identication of potential precursors of myobroblasts. PLoS One. 2010;5:e12949. Sefton MV, Babensee JE, Woodhouse KA. Innate and adaptive immune responses in tissue engineering. Semin Immunol. 2008;20:8385. Badylak SF, Gilbert TW. Immune response to biologic scaffold materials. Semin Immunol. 2008;20:109116. Jones KS. Effects of biomaterial-induced inammation on brosis and rejection. Semin Immunol. 2008;20:130136. Anderson JM, Rodriguez A, Chang DT. Foreign body reaction to biomaterials. Semin Immunol. 2008;20:86100.

104.

105.

106. 107.

108. 109. 110.

111.

112.

113.

114.

115.

116.

117.

118.

119.

120. 121. 122. 123.

20
124. Zhu J. Bioactive modication of poly(ethylene glycol) hydrogels for tissue engineering. Biomaterials. 2010;31:4639 4656. 125. Lee JY, Choi YS, Lee SJ, Chung CP, Park YJ. Bioactive peptide-modied biomaterials for bone regeneration. Curr Pharm Des. In press. 126. Hersel U, Dahmen C, Kessler H. RGD modied polymers: biomaterials for stimulated cell adhesion and beyond. Biomaterials. 2003;24:43854415. 127. Nie T, Akins RE Jr, Kiick KL. Production of heparin-containing hydrogels for modulating cell responses. Acta Biomater. 2009;5:865875. 128. Hsiong SX, Boontheekul T, Huebsch N, Mooney DJ. Cyclic arginine-glycine-aspartate peptides enhance three-dimensional stem cell osteogenic differentiation. Tissue Eng Part A. 2009; 15:263272. 129. Reem T, Tsur-Gang O, Cohen S. The effect of immobilized RGD peptide in macroporous alginate scaffolds on TGFbeta1-induced chondrogenesis of human mesenchymal stem cells. Biomaterials. 2010;31:67466755. 130. Chandler EM, Berglund CM, Lee JS, Polacheck WJ, Gleghorn JP, Kirby BJ, Fischbach C. Stiffness of photocrosslinked RGD-alginate gels regulates adipose progenitor cell behavior. Biotechnol Bioeng. 2011;108:16831692. 131. Dee KC, Andersen TT, Bizios R. Design and function of novel osteoblast-adhesive peptides for chemical modication of biomaterials. J Biomed Mater Res. 1998;40:371377. 132. Rezania A, Healy KE. Biomimetic peptide surfaces that regulate adhesion, spreading, cytoskeletal organization, and mineralization of the matrix deposited by osteoblast-like cells. Biotechnol Prog. 1999;15:1932. 133. Lutolf MP, Lauer-Fields JL, Schmoekel HG, Metters AT, Weber FE, Fields GB, Hubbell JA. Synthetic matrix metalloproteinase-sensitive hydrogels for the conduction of tissue regeneration: engineering cell-invasion characteristics. Proc Natl Acad Sci USA. 2003;100:54135418.

Biotechnol. Prog., 2012, Vol. 28, No. 1


134. Huebsch N, Mooney DJ. Inspiration and application in the evolution of biomaterials. Nature. 2009;462:426432. 135. Discher DE, Janmey P, Wang YL. Tissue cells feel and respond to the stiffness of their substrate. Science. 2005; 310:11391143. 136. OBrien FJ. Biomaterials and scaffolds in tissue engineering. Mater Today. 2011;14:8895. 137. Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nat Biotechnol. 2005;23:4755. 138. Stevens MM. Biomaterials for bone tissue engineering. Mater Today. 2008;11:1825. 139. Bohner M. Resorbable biomaterials as bone graft substitutes. Mater Today. 2010;13:2430. 140. Boccaccio A, Ballini A, Pappalettere C, Tullo D, Cantore S, Desiate A. Finite element method (FEM), mechanobiology and biomimetic scaffolds in bone tissue engineering. Int J Biol Sci. 2011;7:112132. 141. Sreejalekshmi KG, Nair PD. Biomimeticity in tissue engineering scaffolds through synthetic peptide modications-altering chemistry for enhanced biological response. J Biomed Mater Res A. 2011;96:477491. 142. Sun HH, Jin T, Yu Q, Chen FM. Biological approaches toward dental pulp regeneration by tissue engineering. J Tissue Eng Regen Med. 2011;5:e1e16. 143. Evans CH, Palmer GD, Pascher A, Porter R, Kwong FN, Gouze E, Gouze JN, Liu F, Steinert A, Betz O, Betz V, Vrahas M, Ghivizzani SC. Facilitated endogenous repair: making tissue engineering simple, practical, and economical. Tissue Eng. 2007;13:19871993. 144. Quaglia F. Bioinspired tissue engineering: the great promise of protein delivery technologies. Int J Pharm. 2008;364:281297. Manuscript received May 12, 2011, and revision received July 11, 2011.

Das könnte Ihnen auch gefallen