The use of bioactive proteins and peptides to promote osseointegration and success of dental implants: a narrative review
Introduction
The advent of dental implants has revolutionized the surgical and restorative aspects of dentistry, as they have become an excellent and more predictable alternative to fixed and removable prostheses. Although the success of dental implants has been shown to be rather high with survival rates of 93–97% (1), the placement of dental implants in clinical practice has unfortunately led to many post-operative complications, which include inadequate osseointegration and peri-implantitis (2). Although these conditions have led to implant failure, the literature has proposed ways to mitigate these mechanisms and/or slow their progression. These include promoting the quality of the bone-implant interface, accelerating bone formation surrounding implants, inhibiting bacterial adherence, etc. (3-6). The literature has demonstrated many ways that biological proteins and peptides can be used for the purposes mentioned above. The use of these molecules for promoting implant success is a very promising strategy due to high compatibility between proteins and the immune response, ease of synthesis, high antibacterial activity, and bone forming properties (7). However, data regarding the use of proteins and peptides in the field of implantology is relatively scattered. Concise results compiling the relevant molecules into one study would be of high value to the clinician or institution that practices surgical implant placement. Our objective for this study is to review the major biologic proteins and peptides that have been used to promote the success and osseointegration of medical and dental implants. By summarizing this information, we can provide researchers and clinicians valuable information regarding molecules that have been most heavily studied and show the most promising data for promoting implant success. We expect proteins and peptides to be key parts of the arsenal that clinicians use to treat patients needing dental implants, and can hopefully save many patients from experiencing implant failure. We present this article in accordance with the Narrative Review reporting checklist (available at https://fomm.amegroups.com/article/view/10.21037/fomm-24-25/rc).
Methods
The methodology of this literature review was inspired by Jurczak et al., 2020 (7) review of proteins, peptides, and peptidomimetics in implant surface functionalization. Analysis of this study revealed that three broad categories of proteins and peptides exist in the use of implant surfaces: (I) extracellular matrix (ECM) components; (II) growth factors; and (III) antimicrobial peptides (AMPs). Based on this, our search engine strategy involved 3 separate keyword searches into Google Scholar, PubMed, and MEDLINE searches. Each search contained the keywords “implant”, “osseointegration”, “peri-implantitis”, and the 3 different searches differed in the addition of “extracellular matrix and growth factors”, “antimicrobial peptides”, and “proteins and peptides” within the years 2012–2021. There were 9 total searches with the inclusion of Google Scholar, PubMed, and MEDLINE (Table 1). The search yielded 4,906 results total and relevant studies were analyzed that fit our inclusion criteria. Our inclusion criteria consisted of studies that tested the effects of biologic proteins and peptides on the biologic interactions between host cells and implant surfaces/materials in vitro and in vivo, including those belonging to both medical and dental implants. No language considerations were used for our inclusion and exclusion criteria. Host cells included mesenchymal stem cells, osteoblasts, epithelial cells, and other cells involved in promoting implant integration. Implant surfaces included those belonging to orthopedic and dental implants of either ceramic, metal, or polymeric. We excluded studies that did not fit this criteria. We also utilized backward citation searching to obtain 7 additional studies that were published earlier than 2012. The authors each reviewed the articles and analyzed the studies independently. The primary goal of our analysis was to extract data regarding what molecules have shown promising results regarding osseointegration of implants. These studies were highly relevant and provided additional insight into the protein/peptide of interest.
Table 1
Items | Specification |
---|---|
Date of search | 03/2022 |
Databases and other sources searched | PubMed, MEDLINE, Google Scholar |
Search terms used | “Implant” AND “osseointegration” AND “peri-implantitis” OR “extracellular matrix” OR “growth factors” OR “antimicrobial peptides” OR “proteins and peptides” |
Timeframe | 2012–2021 |
Inclusion criteria | Include studies that tested the effects of biologic proteins and peptides on the biologic interactions between host cells and implant surfaces/materials in vitro and in vivo of dental and medical implants |
Selection process | The selection process was independently conducted by J.D.L. and G.E.R. |
Additional consideration | Backward citation searching utilized to obtain 7 additional studies prior to 2012 |
Our search engine strategy yielded a total of 106 studies to be reviewed. All 106 studies could be divided into four sections: (I) ECM components; (II) growth factors; (III) AMPs; and (IV) miscellaneous. The miscellaneous catergory involved a diverse array of proteins and peptides that did not quite fit into the above categories. The majority of molecules in this study belonged largely to the ECM category, with 32 studies investigating 14 different proteins/peptides. The following sections will summarize the functions of these categories of molecules as well as the findings associated with their effects on implant surfaces and cellular responses.
Mechanisms of incorporation
According to the literature, it seems as though there are four main ways proteins and peptides are utilized for promoting implant integration: coating onto implant surface, release systems, local injection, and systemic administration. Out of the studies we analyzed, the most common technique was coating the molecule of interest onto the implant surface. Immobilization of a moeclue onto an implant surface involves chemical tethering procedures utilizing covalent bonding, including ultraviolet (UV)-mediated crosslinking, silanization, and polyethelyne glycol coating (7). Physical adsorption, which involves the use of non-covalent interactions between the molecule and surface, have also been used as an immobilization method (8). Release systems incorporate specific molecules onto the surface of implants, which allow for controlled and local delivery of the molecule into the surrounding tissues (9). Local injection and systemic administration involve delivery of the molecule without dependence of implant surface modification, as these molecules are injected locally around the implant or systemically via subcutaneous access. Each of these techniques are indicated depending on the molecule of interest and can be associated with certain negative outcomes.
ECM components
Glycoproteins
The ECM is present within all tissues and organs and is composed of non-cellular components such as protein, glycosaminoglycan, and glycoconjugate (10) (role of extracell, Frantz 2010). The importance of the ECM lies in its physical scaffolding for cellular elements and its role in initiating crucial biochemical processes that are required for tissue changes and homeostasis (10). Regarding the fibrous protein component of the ECM, the main molecules of this class are collagens, fibronectins, elastins, and laminins (10). Initial interaction of cells with the implant surface is accomplished through specific receptors such as integrins, which is important for promoting the adhesion of bone cells for osseointegration (11). Integrins mediate cellular binding to matrix proteins, which enables cell-ECM interactions to facilitate differentiation of osteoblasts (11). The molecular and cellular interactions between the ECM and osteoblasts are incredibly important for promoting osseointegration of dental implants, and various studies in the literature have confirmed positive effects of coating ECM proteins onto the surface of implants. A complete list of molecules we have reviewed can be found in Table 2.
Table 2
Protein | Function | Study | In vivo/in vitro | Findings | Reference |
---|---|---|---|---|---|
Type I collagen | Mediator of osteoblastic functions, binds to integrins for attachment and differentiation of osteoblasts | Effect of coating type I collagen to titanium on hMSCs | In vitro | Improved hMSC functions, greater osteogenic activity | (12) |
Growth of stem cells in pores of collagen-Ti coatings | In vitro | Greater proliferation of hMSCs and higher trabecular bone formation | (13) | ||
Cross-linked collagen onto Ti implants | In vivo | Higher osteogenic differentiation and bone volume | (14) | ||
Effect of collagen-coated Ti on HDFs | In vitro | Increased fibroblast activation; enhanced remodeling of matrix components | (15) | ||
GFOGER | Binds to a2b1 integrins | Bioactivity of bone marrow stromal cells following GFOGER coating | In vivo | Enhanced osteoblastic differentiation and mineral deposition in stromal cells | (16) |
P-15 | Enhances cell attachment and matrix formation | Implantation into the tibiae of beagle dogs | In vivo | Higher bone-implant contact | (17) |
Hydroxyapatite-P-15 coatings implanted into adult pigs | In vivo | Higher bone-implant contact | (18) | ||
Anti-inflammatory and anti-catabolic effects of P-15 around articular chondrocytes | In vitro | Increased thickness of pericellular matrix, less inhibition of mesenchymal cells, decreased catabolic events | (19) | ||
Fibronectin | Mediator of cell attachment, growth, and migration. Involved in platelet aggregation and adhesion of integrin receptors | Insertion of fibronectin-coated implants into the mandibular premolar region of beagles | In vivo | Increased bone volume, higher osteoblasts surround implants | (20) |
Effect of fibronectin coating on bioactivity of human gingival fibroblasts | In vitro | Improved cell adhesion, proliferation, and spreading | (21) | ||
Osteoblastic effects of fibronectin-Ti64AlV on MC3T3-E1 cell line | In vitro | Enhanced cellular adhesion and bioactivity | (22) | ||
GRGDSP | Promotion of cell adhesion and upregulation of osteoblast markers | MC3T3-E1 cells cultured on Ti discs were immobilized with GRGDSP | In vitro | Enhanced cell adhesion and proliferation, higher expression of osteoblast markers | (23) |
F20 | Stimulation of adhesion and proliferation of osteoblasts | Implantation of GRGDSP coatings into the mandible of rabbits | In vivo | New bone formation in bone-implant contact area | (24) |
Synthetic F20 coated onto titanium discs with ST2 cells seeded onto the surface | In vitro | Increased cellular proliferation and osteoblast differentiation | (25) | ||
Laminin-1 | Binds to integrins and enhances cell adhesion and migration | Effect of implant coating with laminin-1 on early states of osseointegration in vivo | In vivo | Higher removable torque and bone area | (26) |
Promotes angiogenesis | In vivo analysis of laminin-1 coating in the tibiae of rabbits | In vivo | Higher expression of osteoblast and osteoclast markers | (27) | |
Laminin 332 | Binds to integrins and enhances cell adhesion and migration. Promotes angiogenesis | Observation of Lam 332 and ameloblastin immobilization to Ti substrates on human keratinocytes | In vitro | Increased proliferation of keratinocytes and hemidesmosome formation | (28) |
DLTIDDSYWRI | Mediates cell attachment through syndecan-1 | In vivo outcomes of DLTIDDSYWRI coatings in rabbit tibiae | In vivo | Enhanced collagen deposition and ALP activity, improved osseointegration | (29) |
RNIPPFEGCIWN | Ligand for a3b1 integrin, induces membrane recruitment | Measurement of bone formation in vivo using rat models with Ti implant coating | In vivo | Enhanced bone formation and osteoblastic functions | (30) |
ELRs | Mimicking of extracellular matrix and linkage of bioactive molecules to implant surface | Coating of AMPs to titanium implants utilizing ELR linkage | In vitro | High cytocompatibility between coating and gingival fibroblasts | (31) |
Immobilization to Ti substrates and recording of human mesenchymal cells response and physical adsorption | In vitro | Reduced serum protein absorption and improved mesenchymal cell response | (32) | ||
Coating of ELR and chitosan to titanium implant to observe osteoblastic and anti-bacterial activity | In vitro | Increase mineralization, enhanced pre-osteoblastic response, and potent anti-bacterial activity | (33) | ||
Response of MG63 cells following incorporation of ELP into Ti64Al4V and cp-Ti surfaces. In vivo implantation in rat tibia and femur | In vivo | Rapid adhesion of MG63, higher mineralization, increase ALP activity, higher bone-implant contact | (34) | ||
OPN | Regulation of angiogenesis, cell adhesion and migration. Bind to integrins and enhances bone formation | OPN and OPN-derived peptides coated onto Ti surfaces and placed in a canine model | In vivo | Higher bone-implant contact and surrounding bone density | (35) |
OPN peptide coatings implanted into the maxillae of mice | In vivo | Higher rates of osteogenesis around implants | (36) | ||
Ti implants modified with OPN inserted into tibial diaphysis of minipigs | In vivo | Increased formation of new bone | (37) | ||
BSP | Promotes cell attachment, angiogenesis, and matrix mineralization. Regulates adhesion and differentiation of osteoblasts | Effects of human osteoblasts and mouse fibroblasts after coating BSP to titanium surfaces | In vitro | Increased cell migration, mineral deposition and RUNX2 expression | (38) |
BSP-coated scaffolds implanted into lateral femoral condyles | In vivo | No significant effect | (39) | ||
Whether synthetic scaffolds coated with BSP could enhance in vitro osteogenic differentiation and in vivo bone formation | In vivo | No enhancement of in vivo or in vitro osteoconductive properties | (40) | ||
DMP1 | Promotes bone matrix formation and dentin mineralization | MG63 cellular activity due to coating of DMP1 onto titanium | In vitro | Better in vitro bioactivity | (41) |
Effects of DMP1-coated discs on hMSC activity | In vitro | Greater cell proliferation and cellular density, higher alkaline phosphate activity and mineral deposits, increased expression of osteoblast markers | (42) | ||
Bone regeneration and osseointegration of implants coated with DMP1 in maxillary sinus floor of dogs | In vivo | Increase in compact radiopaque area, bone mineral density, and bone-implant contact | (43) | ||
RGD | Integrin-binding domain that promotes cellular attachment to biomaterials | Effects of RGD on Ti surfaces on cell adhesion and osteoblast differentiation | In vitro | Chemical grafting of RGD on Ti enhanced cell adhesion and increased osteoblast markers | (44) |
Tetrapeptide RGD coating on Ti implants investigated for osseointegration | In vitro | Increase in cell growth and higher red blood cell accumulation with fibrin formation | (45) |
Ti, titanium; hMSCs, human mesenchymal stem cells; AMPs, antimicrobial peptides; HDFs, human dermal fibroblasts; ALP, alkaline phosphatase; ELRs, elastin-like recombinamers; OPN, osteopontin; BSP, bone sialoprotein; DMP1, dentin matrix protein 1; RGD, Arg-Gly-Asp.
Type I collagen is the most abundant protein in the ECM and is a mediator of osteoblastic functions (46). Many studies involving titanium implants have demonstrated the ability for type I collagen to regulate osteogenic activity, promote bone formation, and activate fibroblasts (12-15). Other than type I collagen, other glycoproteins such as fibronectin, laminins, and derivatives of elastin can influence implant dynamics. Fibronectin is a large glycoprotein that can be found in various tissues throughout the body and is crucial in cell-ECM interactions and can participate in platelet aggregation (47,48). Studies have shown fibronectin to enhance the cellular responses surrounding titanium implant surfaces in vitro and in vivo (20-22,49). Similar findings are seen in other matrix proteins such as laminins (26,27) and elastin-like polypepides (31-34). Laminins are glycoproteins that are present in the basal lamina of epithelia (50,51) and are capable of binding integrins, which promote adhesion, angiogenesis, and differentiation, which explains many of the positive responses seen in the literature. On the other hand, elastin, which is found in elastic tissue such as blood vessels and ligaments, has been shown function in anchoring AMPs to the titanium implant surface (31), as well as potentiate biomineralization and osteoblastic responses (33).
Although many of these matrix proteins have been shown to be quite successful in promoting bone formation and cell adhesion around implants, many studies have opted to use smaller peptides derived from these proteins in promoting implant success. There are a few reasons as to why peptides offer benefit to larger proteins. For one, full-length matrix proteins (i.e., type I collagen) are often limited by low specificity for particular integrins which can lower the control for cellular responses (16). Also, large matrix proteins often have binding sites for other ligands, which may trigger signaling cascades that can interfere with healing (16). Peptides derived from collagen, such as P-15 and GFOGER, are beneficial due to their smaller sequence that allows for inclusion of only the cell binding sequence of collagen, which limits immunogenicity (52). Peptides derived from type I collagen (P-15, GFOGER), fibronectin (GRGDSP, F20) and laminins (DLTIDDSYWRYI, RNIPPFEGCIWN, laminin 332) also have similar positive outcomes in titanium implant coatings (16-19,23-25,28-30).
SIBLING proteins
SIBLING proteins are a family of non-collagenous glycoproteins in the ECM consisting of osteopontin (OPN), bone sialoprotein (BSP), dentin matrix protein 1 (DMP1), dentin sialophosphoprotein (DSPP), and matrix extracellular phosphoglycoprotein (MEPE) (53). SIBLING proteins share many structural characteristics and are located primarily in dentin and bone. Evidence has shown that SIBLING proteins are involved in matrix mineralization (53), and the coating of SIBLING proteins to the surface of implants have shown promising results.
The SIBLING proteins that have been heavily studied in the literature regarding promoting implant success are OPN, BSP, and DMP1. These proteins have many functions including angiogenesis, cell adhesion, and matrix mineralization (54). OPN has been shown to promote osteogenesis around the implant surface (35-37). The success of OPN coatings is due to its various binding sites for integrins, hydroxyapatite (HA) crystals, collagen, and calcium ions (36), which allows for the deposition of bone following attachment to the implant surface and promotion of osseointegration. DMP1 is another SIBLING protein that has been studied regarding implant surface coatings. Although there are currently no in vivo studies with DMP1, studies have shown that DMP1 can induce osteoblast activity and increase bone mineral density around Ti discs (41-43). This ability of DMP1 can be explained by its multiple phosphorylation sequences and integrin bindings sites that allow it to promote nucleation of calcified matrices (55). BSP, on the other hand, has had some conflicting results in the literature. BSP can promote cellular attachment and differentiation of osteoblasts due to its ability to bind to vitronectin receptors of osteoblasts (56). Although some studies were able to show that BSP coatings increased osteoblast activity and mineral deposition surrounding titanium surfaces (38,57), other studies did not show this ability with other biomaterials such as in calcium phosphate cement (39) and polymer-based scaffolds (40).
Arg-Gly-Asp (RGD)
RGD is a principal integrin-binding domain that is found within matrix proteins such as fibronectin, fibrinogen, OPN, and BSP (58). RGD has been extensively studied in the literature and is highly effective at promoting cell attachment to a variety of biomaterials (58-60). Some advantages of using synthetic RGD are that it can bind to multiple integrin receptors, it can be maintained throughout the processing and sterilization steps needed for synthesis, there is minimal risk of immune reactivity, and it is a relatively inexpensive peptide (58). Interesting results have been noted for the use of RGD in implant coatings. Not only have RGD-immobilized Ti substrates been shown to increase cellular adhesion and proliferation, but can also increase the level of integrins, type I collagen, and sibling proteins around the implant to further enhance osteogenesis (44). Other studies have also shown that type I collagen rich in RGD led to higher fixation speed between the implant and surrounding bone, faster osseointegration and differentiation, and ECM secretion (45,61,62).
Growth factors
Growth factors are biological molecules that are secreted to affect cellular growth, including the promotion or inhibition of cellular division and differentiation (63). Growth factors are either peptides that bind to cell-surface receptors and initiate a downstream cascade of molecular events or are lipid-soluble and bind to intracellular receptors to regulate gene expression (63). They have many important roles in the body, such as in inflammation, wound healing, angiogenesis, and growth (63). Due to these important roles, various growth factors have been analyzed for the incorporation onto dental and medical implants to promote faster wound healing and osseointegration. A complete summary of growth factors can be found in Table 3.
Table 3
Growth factor | Function | Study | In vitro/in vivo | Findings | Reference |
---|---|---|---|---|---|
BMPs | Activate SMAD pathway which leads to differentiation of MSCs into cartilage and bone forming cells | – | – | – | – |
BMP-2 | – | Slow release of BMP-2 from calcium phosphate coating in maxillary sites of minipigs | In vivo | Peak ostoegenic activity and greater newly formed bone | (64) |
BMP-2 and hGDF-5 on the efficacy of osseointegration on MC3T3-E1 cells in vitro and tibiae of rabbits in vivo | In vivo/in vitro | Increaased bone remodeling and bone formation | (65) | ||
Evaluation of in vitro efficacy of BMP-2 on bone-marrow derived mesenchymal stem cells, and in vivo effects of BMP-2 in the tibiae of rabbits | In vivo/in vitro | Increased levels of glycosaminoglycans, osteopontin, and ALP activity. Enhanced bone formation surrounding implant | (66) | ||
BMP-7 | Cellular response of human mesenchymal stem cells as a result of BMP-7 coated titanium | In vitro | Increased cell adhesion, proliferation, mineralization, and osteogenic markers | (67) | |
BMP-7 on osteoblast maturation and extracellular matrix mineralization in vitro. In vivo effects of BMP-7 following implantation in rabbit femurs | In vivo/in vitro | Enhanced osteogenic maturation, increased ECM mineralization, greater thickness of newly formed bone | (68) | ||
BMP-9 | MC3T3-E1 cells in the presence of BMP-9 | In vitro | Expression increases in RUNX2, osterix, BSP, ALP, and osteocalcin. Increased matrix mineralization | (69) | |
FGF-2 | Enhances mitogenic and angiogenic activities in epithelial and bone tissue | FGF-2 coating with titanium and heparin to observe osteoinductivity | In vitro | Increased stability of FGF-2, increased mitogenic and osteogenic pattern in composite layers | (70) |
Percutaneous implantation of FGF-2-coated titanium implants in rabbits | In vivo | Higher bone apposition rate | (71) | ||
In vivo effects of FGF-2 coating with titanium | In vivo | Infection resistance and greater fixation strength | (72) | ||
Analysis of bioactivity in tibiae of rabbits | In vivo | High mitogenic activity and wound healing rate | (73) | ||
VEGF | Increases vascular permeability and endothelial cell migration | Delivery of VEGF to chitosan implant to support osteoblast growth in vitro | In vitro | High alkaline phosphatase activity and calcium deposition | (74) |
Induction of MSCs to endothelial cells in vitro and implantation in back muscles of rats using VEGF-HA scaffolded implants | In vivo/in vitro | Increased expression of endothelial cell markers, increased vessel formation | (75) | ||
Dental implant surfaces enriched in VEGF to improve osteogenic properties of MSCs | In vitro | Protection of aging cells from ROS damage, improved osteogenic and endothelial differentiation | (76) | ||
In vivo analysis of Ti alloy scaffold with VEGF on bone formation and angiogenesis in rabbits | In vivo | Enhanced angiogenesis-mediated bone regeneration and integration | (77) | ||
OGP | Enhances density of newly formed bone and stimulates bone healing | Cellular effects of OGP and fibronectin coated onto Ti implants | In vitro | Increased cellular attachment, MSC proliferation, and osteogenic differentiation | (78) |
Bifunctional coating of OGP and N-acetylcysteine to regulate osteoclast and osteoblast activity | In vitro | Inhibited secretion of inflammatory markers | (79) | ||
Surface modification of PEEK implants and its influence on osteoblast cells in vitro and bone formation in tibiae of rats | In vivo/in vitro | Enhanced attachment, proliferation, ALP activity, and mineralization of pre-osteoblasts | (80) | ||
Biological effects of OGP and ciprofloxacin coating on the ti implant surface | In vitro | Enhanced bone formation surrounding implants; increased osteoblast spreading and differentiation, improved antibacterial activity in vitro | (81) |
hGDF-5, human growth factor and differentiation factor-5; BSP, bone sialoprotein; MSCs, mesenchymal stem cells; HA, hydroxyapatite; Ti, titanium; PEEK, polyetheretherketone; ALP, alkaline phosphatase; OGP, osteogenic growth peptide; ROS, reactive oxygen species; VEGF, vascular endothelial growth factor; FGF-2, fibroblast growth factor-2; BMP, bone morphogenic protein; ECM, extracellular matrix.
Bone morphogenic proteins (BMPs)
BMPs are growth factors that are part of the transforming growth factor-beta (TGF-b) superfamily and play a role in promoting osteogenesis (82). BMPs regulate the bone forming process by binding to BMP receptors and activating the SMAD pathway, which in turn leads to differentiation of mesenchymal cells into cartilage and bone forming cells (83). BMPs are often utilized for bone graft material, which contributes to higher bone to implant contact (BIC) (84). The use of BMPs on the surface of implants have been reported to improve osteogenesis, osteoblast activity, chondroblast activity, and osseointegration (84). Out of the 15 total BMPs, the most effective for inducing bone morphogenesis following surgical implantation are BMP-2 and BMP-7, and to a lesser extent BMP-9 (84-86). BMP-2 is the peptide with the most evidence of promoting osseointegration. Studies have shown that administration of BMP-2 on the surface of Ti discs and implants leads to increased osteoblast activity, glycosaminoglycans, and bone remodeling (64-66). BMP-7 has also been reported to enhance osteogenic potential both in vitro and in vivo (67,68). Although it is worth noting that a study done by (87) did not find any effect of BMP7 on implant attachment. BMP-9 has been the least studied in the family, but has been shown to exhibit even higher osteogenic properties compared to BMP-2 and BMP-7 (85,86). A study done by Souza et al., 2018 (69) observed the effects of BMP-9 on osteoblast differentiation following administration to titanium surfaces. The results showed overall increases in osteoblastic markers, however more research is needed to confirm the effects of BMP-9 on implant surfaces.
Fibroblast growth factor-2 (FGF-2)
FGF-2 has been shown to regulate many cellular processes involving cell proliferation and angiogenesis. The FGF family of proteins are signaling proteins that bind heparin and have high mitogenic and angiogenic activities in tissues including epithelial, connective, bone, and nervous tissue (88). The growth enhancing effects of FGF-2 have led to many studies in the literature testing its effects on titanium surfaces in vivo and in vitro. Positive outcomes have been shown for FGF-2, as studies have shown increases in bone apposition, wound healing, osteogenesis, infection resistance, and bone fixation strength (70-73). It is important to note that heparin can be used to enhance the biological effects of FGF-2, as heparin binds to FGF-2 and can stabilize its action in in vitro (70).
Vascular endothelial growth factor (VEGF)
VEGF is an important growth factor in angiogenesis as it increases vascular permeability and increases endothelial cell migration through chemotaxis (77). Being that blood vessels play an essential role in promoting regeneration and bone repair, insufficient blood supply to areas after bone injury can lead to poor healing (77). Therefore, VEGF is widely used to promote vascular remodeling, and several studies have shown that VEGF is essential for regeneration and bone formation (77,89,90).
Studies have demonstrated promising results with the use of VEGF around the surface of Ti implants (74-77). VEGF has been shown to promote enhanced alkaline phosphatase (ALP) activity and calcium deposition when coated to Ti implants (74). Increased endothelial cell markers such as CD31, FIK-1, and von Willebrand factor (vWF) have been demonstrated in in vivo when VEGF is coated to porous hydroxyapatite scaffolds (75).
Osteogenic growth peptide (OGP)
OGP is a short tetra decapeptide that is identical to the C-terminal amino acid sequence 89–102 of histone H4 (80). OPG has been shown to promote density of newly formed bone and stimulate bone healing (91). Studies have demonstrated the use of OGP on titanium and found osteogenic capabilities (79-81,92). These included enhanced cellular attachment and osteogenesis (80,92), as well as inhibited secretion of inflammatory markers (79). It is worthy to note that these studies utilized OGP as a bifunctional coating, and some of the results can be attributed to the other peptide used in the coating.
AMPs
Biomaterial-associated infection (BAI) can be caused due to the accumulation of biofilm and the presence of antibiotic resistant bacteria (9,93). In order to combat this, the use of AMPs is extremely efficient due to their antimicrobial activity against antibiotic resistant bacteria and bacteria within biofilms. AMPs are part of the innate immune defense or multicellular and microorganisms, and display antimicrobial activity against bacteria, fungi, and enveloped viruses (9,94). The mechanism of action of AMPs is very broad depending on the family of AMP as well as the specific microbe being targeted. In Riool et al’s review of the use of AMPs in biomedical device manufacturing, they subdivided AMPs based on their method of incorporation onto the surface of implants: (I) AMPs used as contact killing surfaces; and (II) AMPs used as release systems. For contact killing surfaces, AMPs are immobilized on the surfaces of medical devices through chemical techniques, whereas AMPs utilized for release systems dissociate from the implant surface and reach the peri-implant tissues (9). Each technique comes with both advantages and disadvantages. Using AMPs through immobilization on the implant surface is highly dependent on the orientation of the covalently linked AMPs as well as the chemical tethering procedure, meaning that using AMP coatings may have significant reduced bactericidal compacity compared to the peptide in the free form (9,95). Another disadvantage also implicated is the inability for the AMP coatings to extend further out into the peri-implant tissues, as these coating are localized to the implant surface (9). On the other hand, utilizing AMPs through releasing mechanism can cause a selection for resistant bacteria and are also potentially ineffective against intracellular bacteria (9). It is therefore suggested that the best use of AMPs on medical devices involves synthesizing peptides with broad spectrum activity with no development of resistance, and tethering these AMPs to biomaterial surfaces along with mechanisms to control its release into the environment (9).
There are a few significant AMP families that have been studied in the literature regarding its use on medical devices. These include human defensins, histatins, and LL-37. There are also many other AMPs that do not fit these categories but have also been studied for similar purposes. A full list of AMPs included in this review can be found in Table 4. Human beta defensins (HBDs) have many roles in the immune system such as bacterial, fungal, and viral killing (120) as well as keratinocyte differentiation (121) and tissue remodeling (122,123). HBDs are highly expressed and rapidly induced at epithelial surfaces in response to infection (112,124,125), and these peptides have been incorporated onto implant surfaces to prevent bacterial accumulation (99,111). Results from these studies show that HBDs (HBD-2) coated onto implants have highly efficient antimicrobial activity and can prevent the colonization of both gram-positive and negative bacteria (111,113), and can also increase the survival of keratinocytes and osteoblasts (112). LL-37 and histatins have also shown to promote osteogenic differentiation, adhesion, and antibacterial activity upon incorporation to the implant surface (114-119). LL-37 is a member of the cathelicidin family, and its function involves the promotion of wound healing and neutralization of lipopolysaccharide (LPS) (126), whereas histatins are found in saliva and are shown to exhibit potent antiungal, antibacterial, and antiviral capabilities (127).
Table 4
Antimicrobial peptide | Function | Study | In vitro/in vivo | Findings | Reference |
---|---|---|---|---|---|
Bacitracin | Bactericidal activity against gram positive organisms. Inhibits cell wall synthesis (pharmacology of specific drug groups) | Immobilization of bacitracin to Ti surfaces for implant integration and anti-bacterial action in rats | In vivo | Implant coating exhibited prophylaxis potential and improved osseointegration | (96) |
GL13K | Derived from salivary BPIFA2, inhibits gram negative bacterial action | GL13K coating to prevent S. gordonii infections under oral cavity conditions | In vitro | Cell wall rupture of S. gordonii | (78) |
In vitro outcomes of F. nucleatum and P. gingivalis following GL13K immobilization to Ti surfaces | In vitro | Eradication of F. nucleatum and P. gingivalis growth, high biocompatibility | (97) | ||
hLF-11 | Derived from human lactoferrin, modulatory effect on host immune cells (van der doses) | Efficacy of hLF-11 coatings to reduce burden of S. sanguinis and L. salvivarius | In vitro | Significant reduction in bacterial populations, decrease in bacterial attachment | (98) |
Chimeric peptides | Peptides connecting sequences with antimicrobial functions to sequences that bind Ti | Effects of chimeric peptide immobilization on S. oralis, S. gordonii, and S. sanguinis | In vitro | Strong antibacterial activity and inhibition of streptococci biofilm | (99) |
Effects of chimeric peptide coatings on Streptococcus spp. | In vitro | High antibacterial activity | (100) | ||
In vitro efficacy of chimeric peptide coatings on Streptococcus spp. and E. coli | In vitro | Significantly reduced adhesion of S. mutans and S. epidermidis | (101) | ||
SESB2V | Bactericidal activity against S. aureus and P. aeruginosa | Functionalized Ti with SESB2V in a rabbit keratitis model for potential application of corneal skirts | In vitro | Lower incidence and decreased extent of infection | (102) |
Melimine | Broad spectrum antibiotic against bacteria, fungi, and protozoa | Melimine coating to reduce in vitro bacterial adhesion and in vivo bacterial load in rodent models | In vivo/in vitro | Reduction of adhesion and biofilm formation of P. aeruginosa and reduction of bacterial load in vivo | (103) |
GZ3.27 | Lipopeptide with antibacterial capabilities against S. aureus and P. aeruginosa | In vitro potency of GZ3.27c upon immobilization to surfaces | In vitro | Damage to cellular envelopes of P. aeruginosa and E. coli | (104) |
Tet20 | Membrane active peptides with antibacterial effects | Effects of Tet20 coatings on antimicrobial activity | In vitro/in vivo | Enhanced antimicrobial activity | (105) |
Te213 | Effects of Tet213 coatings on S. aureus populations in vitro | In vitro | Significant biofilm reduction | (106) | |
OP-145 | Derived from LL-37, reduces growth of multi-resistant strains | In vitro analysis on the ability of OP-145 to inhibit S. aureus. In vivo implantation in rabbit model to observe biofilm inhibition | In vitro/in vivo | Highly effective killing of S. aureus, reduced number of positive cultures around implants | (107) |
Cateslytin | Activates histamine release and displays antimicrobial activity against wide range of organisms | Antimicrobial action of cateslytin coating | In vitro | Potent antimicrobial activity against P. gingivalis, inhibition of bacterial metabolic activity and viability | (108) |
HHC-36 | Cationic peptide with potent antimicrobial properties and low resistance | Culturing of hMSCs with ti-coated HHC-36 | In vitro | Enhanced adhesion, antimicrobial action, and osteogenesis | (109) |
SAAP-145/SAAP-276 | Membrane permeabilization of MRSA strains | Incorporation of SAAP-145/276 into PLEX coating to reduce biomaterial associated infection | In vivo | Reduction in culture positive implants, reduction in MRSA numbers and tissue colonization | (110) |
Human defensins | Protection of epithelial cells. Bacterial, fungal, and viral killing, keratinocyte differentiation and tissue remodeling | – | – | – | – |
HBD-2 | Incubation of HBD-2 functionalized ti implants with E. coli | In vitro | 100% killing rate of E. coli | (111) | |
HBD-2 on the proliferation and survival of osteoblasts, keratinocytes, and MSCs | In vitro | Increased proliferation of MSCs, osteoblasts, and keratinocytes | (112) | ||
RRRRRGALARRRRRRDALAG | Dual coating based of HBD sequence and thiol-functionalized HA to prevent colonization at the implant surface | In vitro | Short term release of peptide, long term stability of AMP presence | (113) | |
HBD-3 | Incorporation of sequences derived from HBD-3 with TiBPs to observe effects against Streptococcus spp. | In vitro | Strong antibacterial activity and inhibition of streptococci biofilm | (99) | |
LL-37 | Promotes wound healing, activates adaptive immunity, and neutralizes lipopylsaccharide | Effect on the proliferation and differentiation of MC3T3-E1 cells | In vitro | Enhanced bacterial activity and osteogenic induction | (114) |
MSC migration in vitro and bone formation in rabbit femur in vivo following Ti surfaces modified with LL-37 | In vivo/in vitro | Increased viability, adhesion, migration, and osteogenic differentiation. Significant bone formation following implantation | (115) | ||
Histatins | Exhibit antibacterial properties and promotes attachment, spreading, and adhesion of epithelial cells | ||||
JH8194 | Assessment of peri-implant and trabecular bone levels in dog mandibles | In vivo | Increased trabecular bone levels | (116) | |
Histatin1 (Hst1) | Effect of histatin1 coated implant surfaces on oral epithelial cells and fibroblasts | In vitro | Enhanced spreading and attachment of oral epithelial cells and fibroblasts to Ti and HA | (117) | |
Effect of Hst1 on osteogenic cells in vitro | In vitro | Enahnced spreading of osteogenic cells | (118) | ||
Celluar responses of Hst1 on Ti discs | In vitro | Higher cell attachment, proliferation, and osteogenesis-related gene expression | (119) |
Ti, titanium; hMSCs, human mesenchymal stem cells; PLEX, polymer-lipid encapsulation matrix; MRSA, methicillin-resistant Staphylococcus aureus; TiBPs, titanium binding peptides; HA, hydroxyapatite; MSC, mesenchymal stem cell; HBD, human beta defensin.
Other proteins and peptides
Animal-inspired peptides
The utilization of peptides derived from animals have been widely used for biomedical applications, as they are often beneficial due to their mechanical properties, biocompatibility, and chemical interactions (128,129). Mainly, for the application of medical implants, peptides have been obtained or derived from arthropods such as Bombyx mori, Anthrea pernyi and spiders Nephila claveips and Araneus diadematus as well as marine mussels. Notably, silk fibroin (SF) and peptides derived from mussels have shown the most evidence of promoting implant success in the literature. The benefits of SF are due to its flexibility, self-assembling ability, biocompatibility, and mechanical properties (130). It is also a low-cost protein extracted from the cocoons of silkworms, making it a widely accessible option. It has also been reported to regulate several pathways responsible for bone remodeling and development (131,132). SF has been extensively tested in the literature as a drug-loaded coating for medical implants, notably in orthopedics (133). Many of the results indicate that the use of SF can promote mineralization, osteoblast adhesion, and osteoblast differentiation at the implant surface, all while minimizing an inflammatory response (134-136).
Peptides derived from the blue mussel Mytilus edulis have been utilized for the coating onto medical devices (137). The power of these peptides lie in the ability of Mytilus edulis to form strong bonding interfaces with various substrates (137), which can be attributed to mussel foot proteins that contain various amounts of lysine and 3,4-Dihydroxy-L-phenylalanine (DOPA) (138). The DOPA component of these peptides are highly important to their ability to self-polymerize and form surface-adherent films onto various materials (139). These peptides shine in their ability to form adherent surfaces that can link various bioactive molecules to the metal surface, such as AMPs, BMPs and RGD (109,140,141). The result is a stable and well controlled utilization of bioactive molecules that can promote antimicrobial and osteogenic activity (109,140). Not only are mussel-inspired peptides useful in forming dual coatings, but they have also been shown to induce osteogenesis on their own when coated onto polymer, metal, and ceramic implant coatings (142).
Titanium-binding peptides (TiBPs)
TiBPs are a group of inorganic peptides that bind to titanium with high affinity and can be conjugated to other molecules to enhance bioactivity (143). Immobilization of bioactive molecules on the implant surface along with TiBPs has been extensively studied, including ECM proteins, RGD, polyethylene glycol (PEG), and BMP (143-146). However, the usefulness of TiBPs stems from its ability to target inorganic substrates (Ti) with high affinity and specificity while interacting with other bioactive molecules, hence serving as an anchor for these molecules to the implant surface. The literature has shown that when TiBPs are incorporated onto the implant surface along with RGD or AMPs, significant osteogenesis and prevention of bacterial colonization were observed (143,147).
Albumin
Albumin accounts for more than 50% of the proteinaceous component of blood and plays a key role as a protein carrier for transportation of various ligands (148). Albumin has been shown to promote adsorption of biomolecules (149-152) as well as hydroxyapatite nucleation (153), which are very important mechanisms for the osseointegration of implants. Likewise, serum albumin has been shown to increase corrosion resistance of Ti6Al4V (154). These features make albumin an attractive candidate for implant coatings to promote successful osseointegration. There have been studies in the literature that have tested these characteristics on titanium and polyetheretherketone (PEEK) implants. These studies have shown that albumin coatings can prevent bacterial adhesion, resist corrosion, and promote osseointegration (149,155-157).
Antibodies
The WNT/β-catenin pathway is involved in the growth and development of many organs and tissue types, particularly in bone and teeth (158). Negative regulators of this pathway, including sclerostin and dickkopf-1 (DKK-1), contribute to marked bone resorption and osteoclast activity. Therefore, inhibiting the action of these negative regulators can inhibit bone loss and enhance bone formation (159). Recent therapeutics to prevent alveolar bone loss following implant surgery involve antibodies to both sclerostin and DKK-1.
Sclerostin is a small glycoprotein that is a potent inhibitor of the WNT/β-catenin signaling pathway, leading to decreased bone formation (160-162). This protein is predominantly expressed by osteoblast lineage cells, and its expression leads to inhibition of bone formation and increased osteoclast activity (163). The hypothesis is that sclerostin antibodies can lead to increased bone formation and decreased bone resorption by blocking the binding of sclerostin to WNT co-receptors, which leads to elevated osteoblastic activity (160). Systemic administration of sclerostin antibodies has been shown to improve bone-implant contact, bone mineral density, and cementogenesis (160,164,165). It is worth to note that local administration of sclerostin antibodies has not been shown to heal periodontal osseous defects (165), which indicates that systemic administration of antibodies should be considered over local to promote osseointegration.
DKK-1 is another inhibitor of bone formation through downregulating the Wnt signaling pathway. It has been reported that mice overexpressing DKK-1 have reduced bone formation and low bone mineral density (166) and DKK-1 upregulation can lead to alveolar bone loss (167) Contrastingly, humans with mutations associated with lowered DKK-1 and sclerostin levels have systemically increased bone mineral density (168). Because of the action of DKK-1, the blockage of DKK-1 activity along with sclerostin has promising results. It has been reported that sclerostin antibody (Scl-Ab) therapy may be limited by reactive increase in DKK-1 expression, which makes DKK-1 antibodies a very attractive strategy to promote bone formation around implants. Results from several studies show that DKK-1 antibodies along with sclerostin antibodies can augment peri-implant bone formation in rodents and non-human primates (159,169).
Hormones
There are three main hormones that have been extensively studied in the literature for the promotion of implant integration: parathyroid hormone (PTH), growth hormone (GH), and melatonin. PTH is produced by the parathyroid glands and plays a role in controlling extracellular calcium and phosphate metabolism (170). PTH can induce bone formation through the downregulation of sclerostin (171). It has been shown that low, intermittent doses of PTH can result in osteoanabolic effects (172) and that PTH may enhance bone formation through the promotion of osteoblast maturation and decreasing osteoblast apoptosis (173-175). Results from various studies show that intermittent administration of PTH is effective at promoting new bone formation around implants (173,176-179). A summary of these hormones can be found in Table 5.
Table 5
Protein/peptide | Function | Study | In vitro/in vivo | Findings | Reference |
---|---|---|---|---|---|
Silk fibroin | Protein extract from cocoons of silkworks. Regulate bone remodeling, highly flexible and biocamptible | AgNPs/gentamycine-loaded structure-controlled silk fibroin coatings developed to promote success of Ti implants | In vivo | Improvement in generation of new-born bone and bonding between bone and tissue in rabbit femoral defect with minimal inflammation | (133) |
Silk fibroin coated onto Ti6Al4V implants with TiO2 nanotubes to improve osteogenic potential | In vitro | Increased adhesion of osteoblasts and hMSCs, increased alkaline phosphatase, increased expression of osteogenic markers | (128) | ||
PEEK implants coated with silk fibroin and bone forming peptide to increase bioactivity and osteogenesis | In vitro | Increased cellular proliferation and adhesion, maturation of osteoblast-like cells | (180) | ||
Mussel inspired peptides | Derived from Mytilus edulis. Lysine and DOPA components promote self-polymeriztion and adherence of bioactive molecules to metal surface | Multifunctional hydrogel coating onto surface of Ti implants to prevent infection and enhance osteogenesis | In vitro | Improved adhesion of AMPs to Ti surface, enhanced osteogenesis of hMSCs | (109) |
Clickable mussel-inspired peptides (DOPA-N3) grafted onto Ti materials | In vivo | Increased adhesion, mechanical stability, and osteointegration | (140) | ||
Bi-functional coating of mussel inspired peptides for functionalization of Ti screws | In vivo/in vitro | In vitro showed improved osteogenesis of osteoporosis-derived mesenchymal cells. In vivo Ti screws showed increased osteogenesis and mechanical stability | (141) | ||
TiBPs | Anchor for bioactive molecules onto Ti surface | Use of TiBP to immobilize RGD to implant surface and enhance osteogenesis of pre-osteoblasts and fibroblasts | In vitro | Effective immobilization of RGD to implant surface, enhanced bioactivity of pre-osteoblast and fibroblast cells | (143) |
Bifunctional peptide composed of TiBPs and AMPs to coat the implant surface | In vitro | 100% binding to titanium, durability to electronic toothbrushes, retention of antimicrobial challenge | (147) | ||
Albumin | Blood protein carrier, promotion of biomolecule adsorption, nucleation of hydroxyapatite, and corrosion resistance | Albumin coating on Ti64Al4V to increase corrosion resistance and control protein adsorption | In vitro | Increased adsorption of amino acids, decreased dissolution of vanadium to environment | (149) |
Investigation of synergistic effects of albumin and peroxide on the corrosion resistance of Ti64Al4V | In vitro | Suppressed dissolution in the presence of hydrogen peroxide at short times | (156) | ||
Albumin nanocarriers coated onto polyethylene implants for long-term growth factor release | In vitro | Prolonged presence of encapsulated small molecules | (157) | ||
Antibodies | |||||
Scl-Abs | Inhibition of sclerostin binding to WNT co-receptors, leading to elevated osteoblast activity | Injection of Scl-Abs following placement of titanium implants in the maxillae of rats | In vivo | Higher bone fill percentage and bone-implant contact | (164) |
Dual effects of zoledronic acid and Scl-Abs following insertion of implants into tibiae of rats | In vivo | Increased bone mineral density, bone volume, and removal torque | (160) | ||
Systemically delivered sclerostin antibodies vs. local administration. Efficacy of antibodies to repair osseous defects surrounding teeth | In vivo | Systemically delivered Scl-Abs increased bone regeneration and cementogenesis | (165) | ||
DABs | Blockage of DKK-1 which activates the WNT signaling pathway and elevates bone formation | Observation of Scl-Abs combined with DABs | In vivo | Scl-Abs combined with DABs exceeded peri-implant bone formation compared to Scl-Ab administration alone | (169) |
Bispecific antibody to sclerostin and DKK-1 to analyze the effects on bone mass in rats, mice, and primates | In vivo | Synergistic bone formation in rodents and non-human primates, increased bone repair | (159) | ||
Hormones | |||||
PTH | Regulation of calcium and phosphate metabolism, induction of bone formation through sclerostin downregulation | Coating of PTH related protein onto HA foams to improve bone repair in aging rats | In vivo | Increased bone volume, trabecular and cortical thickness, increased osteocalcin and VEGF, decreased Sost gene | (176) |
Analyzation of PTH related protein coated with Ti implants on MC3T3-E1 cells in vitro and implant performance in the rat femur in vivo | In vivo | High levels of ALP and expression of osteoblast markers, high bone formation 2 weeks after implantation | (177) | ||
In vitro and in vivo effects of PTH-Ti coatings on BMSCs and in the tibiae of rats | In vitro/in vivo | Increased ALP, BMP-2, Runx2, Osterix in BMSCs. Increase new bone formation and BIC in vivo | (173) | ||
GH | Stimulates release of bone-derived growth factors. Stimulation of collagen, osteocalcin, and ALP synthesis | Topical application of GH on implants in the mandibles of dogs | In vivo | Significant bone formation and inter-thread bone values | (181) |
Topical administration of GH and melatonin in the mandibles and maxillae of dogs | In vivo | High BIC and phosphorous concentration in early stages following surgery | (182) | ||
Placement of recombinant human GH into osteotomy sites prior to implantation in rabbits | In vivo | More abundant bone tissue and accelerate bone repair 2 weeks post-operatively | (183) | ||
Melatonin | Regulation of immune system, inducer of bone formation, inhibitor of bone resorption | Melatonin coated onto implant and its effects on osseointegration in foxhound dogs | In vivo | Improved BIC and bone formation | (184) |
Effects of systemic administration of melatonin on HA-coated titanium implants in osteopenic rats | In vivo | Improved osseointegration, increased BIC, bone volume, trabecular thickness and trabecular number, and increased expression of osteogenesis-related genes | (185) | ||
Coating of melatonin onto Ti substrates and its biological effects on MC3T3-E1 osteoblastic cells and macrophages | In vitro | Increased expression of Runx2 and BMP-2, enhanced adsorption of proteins involved in coagulation and angiogenesis, decreased adsorption of complement | (186) | ||
7ND | Inhibition of CCL2 signaling and reduction of particle induced inflammation and osteolysis | Ti rods coated with 7ND and its effects on peri- implant bone loss | In vitro | Decreased macrophage recruitment, number of osteoclasts, and bone loss | (187) |
Effects of 7ND release from surface of orthopedic implants | In vitro | Reduction of macrophage migration | (188) | ||
Implantation of Ti rods with 7ND coating into the distal femurs of mice | In vivo | No effect | (189) |
Ti, titanium; TiO2, titanium dioxide; PEEK, polyetheretherketone; AgNP, silver nanoparticles; hMSCs, human mesenchymal stem cells; AMPs, antimicrobial peptides; HA, hydroxyapatite; BMSCs, bone marrow mesenchymal stem cells; DOPA, 3,4-dihydrox-L-phenylalanine; TiBP, titanium-binding peptide; VEGF, vascular endothelial growth factor; ALP, alkaline phosphatas; PTH, parathyroid hormone; BIC, bone to implant contact; BMP, bone morphogenic protein; CCL2, motif chemokine ligand 2; 7ND, mutant version of chemoattractant protein-1; DKK-1, dickkopf-1; DAB, DKK-1 antibody; Scl-Ab, sclerostin antibody; RGD, Arg-Gly-Asp.
GH is an anterior pituitary hormone that stimulates the liver to release a variety of bone-derived growth factors (190). GH can have a significant impact on both osteoblast and osteoclast activity (191,192) and it has been suggested that GH enhances new bone formation and increases cortical bone mass (193). Furthermore, GH has also been suggested to stimulate synthesis of collagen, osteocalcin, ALP, and hard tissue mineralization (194,195). In regard to medical implants, it has been shown that GH can increase bone formation and accelerate bone repair in dogs (181-183). However, some limitations have been suggested for the use of GH. Many studies fail to acknowledge the long-term effects of GH administration, which can potentially involve increases in bone resorption (196,197), and there are also inconsistencies among studies regarding the design of the study. It is likely that GH induces bone formation surrounding implants, however studies need to focus on applying GH in a clinical setting as to minimize the conflicting factors that may have influenced the outcomes of other studies (190).
Melatonin is a hormone produced by the pineal gland and other organs including the retina, bone marrow, and intestines (198). The functions of melatonin include control of circadian rhythm and body temperature, regulation of sexual development, and activation of the immune system (199-201). Melatonin has been shown to act as an anti-inflammatory agent, and within the oral cavity can enhance bone formation and reduce bone resorption (198). The literature has shown that melatonin, whether applied topically to the surface of implants or administered systemically, can promote implant success due to its osteogenic, anti-inflammatory, and angiogenic properties in vitro and in vivo (184-186).
7ND
Peri-implant tissue expresses high levels of motif chemokine ligand 2 (CCL2) [monocyte chemoattractant protein-1 (MCP-1)], which plays a key role in the recruitment of macrophages to the site of particle-induced inflammation (189). Blockade of CCL2 signaling can therefore reduce inflammation-induced osteolysis (202), which is another emerging approach to prevent bone loss surrounding implants. 7ND, a mutant version of CCL2, has been shown to inhibit CCL2 signaling (203) and reduce particle-induced inflammation and osteolysis (204,205). Some studies have found that 7ND coatings to titanium rods and orthopedic implants lead to decreased macrophage and osteoclast recruitment (187,188), whereas a study done by Sato 2016 (189) did not observe any effects of the coating. It seems that 7ND has potential to lower inflammation and reduce bone loss around implants, but more studies regarding this protein are needed.
Discussion
To the best of our knowledge, this narrative review is the most comprehensive overview of proteins and peptides that have been studied for promoting osseointegration of dental and medical implants. A previous study written by Jurczak et al., 2020 (7) provides a similar overview of proteins and peptides for the use of implant surface functionalization. However, our study aimed at providing an overview for the clinician and offers a larger list of proteins and peptides that are not limited to surface immobilization. Some strengths of our study are that it includes proteins and peptides utilized via multiple delivery systems. Our study includes molecules used as release systems, immobilized substrates, and as injections, which allows us to evaluate and observe differences in outcomes and discuss pros and cons of each system. Our study also categorizes an overwhelming number of molecules by their mechanisms of action and chemical class, which makes it easier for the reader to comprehend and understand the principles behind this field of study. Limitations of our study included a lack of statistical analysis, as this work is meant to be a narrative review and cannot answer definitive questions regarding the optimal molecule or dispute any controversies that may have arisen in this field. It is also possible that since this study has been conducted, new outcomes of proteins and peptides may have been published but cannot be included in this work due to the dates used in the search criteria. We are well aware that although this is a comprehensive overview, there may be other molecules that have not been discussed which is reasonable due to the rapid advancement of this field and the new studies that are constantly being published.
Based off the studies that have been presented in the literature regarding the use of proteins and peptides to promote implant success, it seems that these molecules can assist in integration through numerous mechanisms, many of which overlap. These include bone formation, mineralization, matrix formation, epithelial proliferation, vascularization, antimicrobial activity, inflammatory inhibition, and anchorage to the implant surface. The overall majority of molecules used as implant coatings consisted of matrix proteins. Matrix proteins such as type I collagen, elastin, and fibronectin are highly beneficial due to their ability to bind integrins and promote osteoblast adhesion to the implant surface, which will lead to their differentiation and bone formation. However, it seems that using smaller peptides derived from these larger proteins is superior because they are faster and cheaper to synthesize, more specific to particular integrins, and have less immunogenicity, and are also easier to incorporate into multifunctional coatings.
Growth factors, on the other hand, have been shown to have benefit over these small peptides (RGD) because these peptides have only led to marginal increases in osseointegration (76) (Zavan). Growth factors are also essential for new tissue production, can be used in regenerative procedures, and can perform feedback controls on inflammatory processes (76). BMPs can directly stimulate osteoblast differentiation and FGF-2 can stimulate not only bone formation but soft tissue formation as well (88). Although growth factors have many positive effects, one limitation of growth factors is their short half-life, particularly limited to a few hours (135). Particularly some growth factors, such as BMP, can lead to long-term adverse effects such as inflammation and bone resorption (206). The aim of using peptides derived from animals (SF) are for their extremely good mechanical properties that can strongly increase implant fixation to bone. They have also been shown to increase osteogenesis and aid in wound healing (133,207).
The use of antibodies to sclerostin and DKK-1 have shown very promising results. By inhibiting a negative regulator of bone formation through the use of antibodies, we can expect to see higher trabecular bone density and osteogenesis surrounding the implant. However, these antibodies have exclusively been shown to work when administered systemically, and adverse effects of using sclerostin antibodies in clinical trials have shown side effects such as cardiovascular incidents (208). Therefore, depending on the duration of treatment, potential adverse effects may be expected as a result of this therapy. Albumin is unique as it can prevent corrosion of titanium implants as well as improve bioactivity of other molecules to the implant surface, and TiBP serve as good anchors for other bioactive molecules. AMPs, although do not possess natural abilities to directly promote bone formation, are very effective at preventing bacterial colonization and infection of medical devices, including orthopedic and dental implants.
Taking all of this into consideration, one can see the many diverse options that exist for using bioactive molecules, particularly proteins and peptides, for the promotion of implant success. An illustration of the mechanisms reviewed can be found in Figure 1. However, based on the literature, it seems the most plausible and logical solution for deciding on the optimal molecule revolves around a coating and/or injection that can utilize all of the elements described above. These include peptides that have high affinity to the titanium surface, can induce differentiation of osteoblasts and promote bone formation, and can prevent bacterial colonization. To the best of our knowledge, no studies have combined molecules that can carry out all of the mechanisms mentioned above. To say that this combination of molecules would be the optimal solution for promoting implant success would require in vivo and in vitro studies to confirm the benefits of such a large combination, followed by clinical trials for mainstream implementation. In relation to clinical practice, many of these molecules differ in chemical structure, interaction with host cells, and potential side effects. Therefore, it may be dependent on the clinician to determine the optimal molecule to incorporate into treatment depending on both the clinical indication and the patient’s medical history, as even though it seems that all of these molecules are efficient in promoting osseointegration in laboratory studies, more clinical trials in humans are needed in order to determine important factors such as adverse effects, optimal dosage, duration of treatment, and drug interferences. Therefore, until large volumes of these studies are carried out, it is not reasonable to conclude what the optimal molecule is for promoting implant osseointegration and preventing peri-implantitis. Recommendations for future studies include in vivo and in vitro studies that test the effects of a combination of molecules that can promote bone formation, exhibit high antibacterial activity, and attach to the implant surface with high affinity. Also, randomized controlled clinical trials testing the effects of any of the molecules discussed in this review are indicated for further investigation.
Conclusions
Many studies seem to indicate that the utilization of proteins and peptides, either through immobilization to implant surfaces or systemic administration, is a promising strategy for preventing implant failure. The mechanisms in which they work is by promoting bone formation, fighting against bacterial infection, and enhancing cellular adhesion to implant surfaces. At this point in time, it is highly likely that the use bioactive molecules under the category of proteins and peptides are efficient at promoting implant success, but more research is needed in humans and randomized clinical trials in order to determine the optimal molecule, combination of molecules, and practical application in order for clinicians to utilize them in practice.
Acknowledgments
This work was presented at the 2024 Academy of Osseointegration Annual Meeting in Charlotte, North Carolina in the form of an eposter.
Funding: None.
Footnote
Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://fomm.amegroups.com/article/view/10.21037/fomm-24-25/rc
Peer Review File: Available at https://fomm.amegroups.com/article/view/10.21037/fomm-24-25/prf
Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://fomm.amegroups.com/article/view/10.21037/fomm-24-25/coif). G.E.R. serves as an unpaid editorial board member of Frontiers of Oral and Maxillofacial Medicine from October 2023 to September 2025. Both authors report that Stony Brook School of Dental Medicine funded the travel and meeting cost for this work to be presented at the Academy of Osseointegration 2024 Annual Meeting in Charlotte, NC. The authors have no other conflicts of interest to declare.
Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.
References
- Albrektsson T, Donos N. Working Group 1. Implant survival and complications. The Third EAO consensus conference 2012. Clin Oral Implants Res 2012;23:63-5. [Crossref] [PubMed]
- Kochar SP, Reche A, Paul P. The Etiology and Management of Dental Implant Failure: A Review. Cureus 2022;14:e30455. [Crossref] [PubMed]
- Wauthle R, van der Stok J, Amin Yavari S, et al. Additively manufactured porous tantalum implants. Acta Biomater 2015;14:217-25. [Crossref] [PubMed]
- Yeo IL. Modifications of Dental Implant Surfaces at the Micro- and Nano-Level for Enhanced Osseointegration. Materials (Basel) 2019;13:89. [Crossref] [PubMed]
- Körtvélyessy G, Tarjányi T, Baráth ZL, et al. Bioactive coatings for dental implants: A review of alternative strategies to prevent peri-implantitis induced by anaerobic bacteria. Anaerobe 2021;70:102404. [Crossref] [PubMed]
- Arciola CR, Campoccia D, Montanaro L. Implant infections: adhesion, biofilm formation and immune evasion. Nat Rev Microbiol 2018;16:397-409. [Crossref] [PubMed]
- Jurczak P, Witkowska J, Rodziewicz-Motowidło S, et al. Proteins, peptides and peptidomimetics as active agents in implant surface functionalization. Adv Colloid Interface Sci 2020;276:102083. [Crossref] [PubMed]
- Sandhyarani N. Chapter 3 - Surface modification methods for electrochemical biosensors. In: Ensafi AA, editor. Electrochemical Biosensors. Amsterdam, The Netherlands: Elsevier; 2019:45-75.
- Riool M, de Breij A, Drijfhout JW, et al. Antimicrobial Peptides in Biomedical Device Manufacturing. Front Chem 2017;5:63. [Crossref] [PubMed]
- Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci 2010;123:4195-200. [Crossref] [PubMed]
- Geissler U, Hempel U, Wolf C, et al. Collagen type I-coating of Ti6Al4V promotes adhesion of osteoblasts. J Biomed Mater Res 2000;51:752-60. [Crossref] [PubMed]
- Ao H, Xie Y, Tan H, et al. Improved hMSC functions on titanium coatings by type I collagen immobilization. J Biomed Mater Res A 2014;102:204-14. [Crossref] [PubMed]
- Ao HY, Xie YT, Yang SB, et al. Covalently immobilised type I collagen facilitates osteoconduction and osseointegration of titanium coated implants. J Orthop Translat 2016;5:16-25. [Crossref] [PubMed]
- Bae EB, Yoo JH, Jeong SI, et al. Effect of Titanium Implants Coated with Radiation-Crosslinked Collagen on Stability and Osseointegration in Rat Tibia. Materials (Basel) 2018;11:2520. [Crossref] [PubMed]
- Marín-Pareja N, Salvagni E, Guillem-Marti J, et al. Collagen-functionalised titanium surfaces for biological sealing of dental implants: effect of immobilisation process on fibroblasts response. Colloids Surf B Biointerfaces 2014;122:601-10. [Crossref] [PubMed]
- Reyes CD, Petrie TA, Burns KL, et al. Biomolecular surface coating to enhance orthopaedic tissue healing and integration. Biomaterials 2007;28:3228-35. [Crossref] [PubMed]
- Coelho PG, Teixeira HS, Marin C, et al. The in vivo effect of P-15 coating on early osseointegration. J Biomed Mater Res B Appl Biomater 2014;102:430-40. [Crossref] [PubMed]
- Lutz R, Srour S, Nonhoff J, et al. Biofunctionalization of titanium implants with a biomimetic active peptide (P-15) promotes early osseointegration. Clin Oral Implants Res 2010;21:726-34. [Crossref] [PubMed]
- Shortt C, Luyt LG, Turley EA, et al. A Hyaluronan-binding Peptide (P15-1) Reduces Inflammatory and Catabolic Events in IL-1β-treated Human Articular Chondrocytes. Sci Rep 2020;10:1441. [Crossref] [PubMed]
- Chang YC, Ho KN, Feng SW, et al. Fibronectin-Grafted Titanium Dental Implants: An In Vivo Study. Biomed Res Int 2016;2016:2414809. [Crossref] [PubMed]
- Kim EC, Lee DY, Lee MH, et al. The Effect of Fibronectin-Immobilized Microgrooved Titanium Substrata on Cell Proliferation and Expression of Genes and Proteins in Human Gingival Fibroblasts. Tissue Eng Regen Med 2018;15:615-27. [Crossref] [PubMed]
- Felgueiras HP, Evans MDM, Migonney V. Contribution of fibronectin and vitronectin to the adhesion and morphology of MC3T3-E1 osteoblastic cells to poly(NaSS) grafted Ti6Al4V. Acta Biomater 2015;28:225-33. [Crossref] [PubMed]
- Pramono S, Pugdee K, Suwanprateep J, et al. Sandblasting and fibronectin-derived peptide immobilization on titanium surface increase adhesion and differentiation of osteoblast-like cells (MC3T3-E1). J Dent Sci 2016;11:427-36. [Crossref] [PubMed]
- Deng YL, Li D, Li MQ, et al. Effect of GRGDSP peptide coating on pure titanium surface on osteointegration. China Journal of Oral and Maxillofacial Surgery 2018;16:199-204.
- Cho YD, Kim SJ, Bae HS, et al. Biomimetic Approach to Stimulate Osteogenesis on Titanium Implant Surfaces Using Fibronectin Derived Oligopeptide. Curr Pharm Des 2016;22:4729-35. [Crossref] [PubMed]
- Bougas K, Jimbo R, Vandeweghe S, et al. Bone apposition to laminin-1 coated implants: histologic and 3D evaluation. Int J Oral Maxillofac Surg 2013;42:677-82. [Crossref] [PubMed]
- Schwartz-Filho HO, Bougas K, Coelho PG, et al. The effect of laminin-1-doped nanoroughened implant surfaces: gene expression and morphological evaluation. Int J Biomater 2012;2012:305638. [Crossref] [PubMed]
- Koidou VP, Argyris PP, Skoe EP, et al. Peptide coatings enhance keratinocyte attachment towards improving the peri-implant mucosal seal. Biomater Sci 2018;6:1936-45. [Crossref] [PubMed]
- Kang HK, Kim OB, Min SK, et al. The effect of the DLTIDDSYWYRI motif of the human laminin α2 chain on implant osseointegration. Biomaterials 2013;34:4027-37. [Crossref] [PubMed]
- Yeo IS, Min SK, Kang HK, et al. Identification of a bioactive core sequence from human laminin and its applicability to tissue engineering. Biomaterials 2015;73:96-109. [Crossref] [PubMed]
- Acosta S, Ibañez-Fonseca A, Aparicio C, et al. Antibiofilm coatings based on protein-engineered polymers and antimicrobial peptides for preventing implant-associated infections. Biomater Sci 2020;8:2866-77. [Crossref] [PubMed]
- Salvagni E, Berguig G, Engel E, et al. A bioactive elastin-like recombinamer reduces unspecific protein adsorption and enhances cell response on titanium surfaces. Colloids Surf B Biointerfaces 2014;114:225-33. [Crossref] [PubMed]
- Govindharajulu JP, Chen X, Li Y, et al. Chitosan-Recombinamer Layer-by-Layer Coatings for Multifunctional Implants. Int J Mol Sci 2017;18:369. [Crossref] [PubMed]
- Raphel J, Karlsson J, Galli S, et al. Engineered protein coatings to improve the osseointegration of dental and orthopaedic implants. Biomaterials 2016;83:269-82. [Crossref] [PubMed]
- Fiorellini JP, Glindmann S, Salcedo J, et al. The Effect of Osteopontin and an Osteopontin-Derived Synthetic Peptide Coating on Osseointegration of Implants in a Canine Model. Int J Periodontics Restorative Dent 2016;36:e88-94. [Crossref] [PubMed]
- Makishi S, Yamazaki T, Ohshima H. Osteopontin on the Dental Implant Surface Promotes Direct Osteogenesis in Osseointegration. Int J Mol Sci 2022;23:1039. [Crossref] [PubMed]
- Aragoneses J, López-Valverde N, López-Valverde A, et al. Bone Response to Osteopontin-Functionalized Carboxyethylphosphonic Acid-Modified Implants. Experimental Study in a Minipig Model. Front Mater 2022;9:914853. [Crossref]
- Baranowski A, Klein A, Ritz U, et al. Surface Functionalization of Orthopedic Titanium Implants with Bone Sialoprotein. PLoS One 2016;11:e0153978. [Crossref] [PubMed]
- Klein A, Baranowski A, Ritz U, et al. Effect of bone sialoprotein coating on progression of bone formation in a femoral defect model in rats. Eur J Trauma Emerg Surg 2020;46:277-86. [Crossref] [PubMed]
- Schaeren S, Jaquiéry C, Wolf F, et al. Effect of bone sialoprotein coating of ceramic and synthetic polymer materials on in vitro osteogenic cell differentiation and in vivo bone formation. J Biomed Mater Res A 2010;92:1461-7. [Crossref] [PubMed]
- Pei X, Pan L, Cui F, et al. The recombinant human dentin matrix protein 1-coated titanium and its effect on the attachment, proliferation and ALP activity of MG63 cells. J Mater Sci Mater Med 2012;23:2717-26. [Crossref] [PubMed]
- Kongkiatkamon S, Ramachandran A, Knoernschild KL, et al. Dentin Matrix Protein 1 on Titanium Surface Facilitates Osteogenic Differentiation of Stem Cells. Molecules 2021;26:6756. [Crossref] [PubMed]
- Ma D, Wang Y, Chen Y, et al. Promoting Osseointegration of Dental Implants in Dog Maxillary Sinus Floor Augmentation Using Dentin Matrix Protein 1-Transduced Bone Marrow Stem Cells. Tissue Eng Regen Med 2020;17:705-15. [Crossref] [PubMed]
- Ryu JJ, Park K, Kim HS, et al. Effects of anodized titanium with Arg-Gly-Asp (RGD) peptide immobilized via chemical grafting or physical adsorption on bone cell adhesion and differentiation. Int J Oral Maxillofac Implants 2013;28:963-72. [Crossref] [PubMed]
- Syam S, Wu CJ, Lan WC, et al. The Potential of a Surface-Modified Titanium Implant with Tetrapeptide for Osseointegration Enhancement. Appl Sci 2021;11:2616. [Crossref]
- Viguet-Carrin S, Garnero P, Delmas PD. The role of collagen in bone strength. Osteoporos Int 2006;17:319-36. [Crossref] [PubMed]
- Lenselink EA. Role of fibronectin in normal wound healing. Int Wound J 2015;12:313-6. [Crossref] [PubMed]
- Cho J, Mosher DF. Enhancement of thrombogenesis by plasma fibronectin cross-linked to fibrin and assembled in platelet thrombi. Blood 2006;107:3555-63. [Crossref] [PubMed]
- Chang YC, Lee WF, Feng SW, et al. In Vitro Analysis of Fibronectin-Modified Titanium Surfaces. PLoS One 2016;11:e0146219. [Crossref] [PubMed]
- Javed F, Al Amri MD, Kellesarian SV, et al. Laminin coatings on implant surfaces promote osseointegration: Fact or fiction? Arch Oral Biol 2016;68:153-61. [Crossref] [PubMed]
- Rohde H, Wick G, Timpl R. Immunochemical characterization of the basement membrane glycoprotein laminin. Eur J Biochem 1979;102:195-201. [Crossref] [PubMed]
- Gomar F, Orozco R, Villar JL, et al. P-15 small peptide bone graft substitute in the treatment of non-unions and delayed union. A pilot clinical trial. Int Orthop 2007;31:93-9. [Crossref] [PubMed]
- Staines KA, MacRae VE, Farquharson C. The importance of the SIBLING family of proteins on skeletal mineralisation and bone remodelling. J Endocrinol 2012;214:241-55. [Crossref] [PubMed]
- Zhao H, Chen Q, Alam A, et al. The role of osteopontin in the progression of solid organ tumour. Cell Death Dis 2018;9:356. [Crossref] [PubMed]
- Srinivasan R, Chen B, Gorski JP, et al. Recombinant expression and characterization of dentin matrix protein 1. Connect Tissue Res 1999;40:251-8. [Crossref] [PubMed]
- Hunter GK, Goldberg HA. Nucleation of hydroxyapatite by bone sialoprotein. Proc Natl Acad Sci U S A 1993;90:8562-5. [Crossref] [PubMed]
- Baranowski A, Klein A, Ritz U, et al. Evaluation of Bone Sialoprotein Coating of Three-Dimensional Printed Calcium Phosphate Scaffolds in a Calvarial Defect Model in Mice. Materials (Basel) 2018;11:2336. [Crossref] [PubMed]
- Bellis SL. Advantages of RGD peptides for directing cell association with biomaterials. Biomaterials 2011;32:4205-10. [Crossref] [PubMed]
- Hennessy KM, Clem WC, Phipps MC, et al. The effect of RGD peptides on osseointegration of hydroxyapatite biomaterials. Biomaterials 2008;29:3075-83. [Crossref] [PubMed]
- LeBaron RG, Athanasiou KA. Extracellular matrix cell adhesion peptides: functional applications in orthopedic materials. Tissue Eng 2000;6:85-103. [Crossref] [PubMed]
- Takeuchi Y, Nakayama K, Matsumoto T. Differentiation and cell surface expression of transforming growth factor-beta receptors are regulated by interaction with matrix collagen in murine osteoblastic cells. J Biol Chem 1996;271:3938-44. [Crossref] [PubMed]
- Göransson A, Jansson E, Tengvall P, et al. Bone formation after 4 weeks around blood-plasma-modified titanium implants with varying surface topographies: an in vivo study. Biomaterials 2003;24:197-205. [Crossref] [PubMed]
- Stone WL, Leavitt L, Varacallo M. Physiology, Growth Factor. Treasure Island (FL): StatPearls Publishing; 2024.
- Hunziker EB, Enggist L, Küffer A, et al. Osseointegration: the slow delivery of BMP-2 enhances osteoinductivity. Bone 2012;51:98-106. [Crossref] [PubMed]
- Yang DH, Moon SW, Lee DW. Surface Modification of Titanium with BMP-2/GDF-5 by a Heparin Linker and Its Efficacy as a Dental Implant. Int J Mol Sci 2017;18:229. [Crossref] [PubMed]
- Pang K, Seo YK, Lee JH. Effects of the combination of bone morphogenetic protein-2 and nano-hydroxyapatite on the osseointegration of dental implants. J Korean Assoc Oral Maxillofac Surg 2021;47:454-64. [Crossref] [PubMed]
- Al-Jarsha M, Moulisová V, Leal-Egaña A, et al. Engineered Coatings for Titanium Implants To Present Ultralow Doses of BMP-7. ACS Biomater Sci Eng 2018;4:1812-9. [PubMed]
- Nemcakova I, Litvinec A, Mandys V, et al. Coating Ti6Al4V implants with nanocrystalline diamond functionalized with BMP-7 promotes extracellular matrix mineralization in vitro and faster osseointegration in vivo. Sci Rep 2022;12:5264. [Crossref] [PubMed]
- Souza ATP, Bezerra BLS, Oliveira FS, et al. Effect of bone morphogenetic protein 9 on osteoblast differentiation of cells grown on titanium with nanotopography. J Cell Biochem 2018;119:8441-9. [Crossref] [PubMed]
- Yasunaga M, Kobayashi F, Sogo Y, et al. The enhancing effects of heparin on the biological activity of FGF-2 in heparin-FGF-2-calcium phosphate composite layers. Acta Biomater 2022;148:345-54. [Crossref] [PubMed]
- Fujii K, Ito A, Mutsuzaki H, et al. Reducing the risk of impaired bone apposition to titanium screws with the use of fibroblast growth factor-2-apatite composite layer coating. J Orthop Surg Res 2017;12:1. [Crossref] [PubMed]
- Mutsuzaki H, Ito A, Sogo Y, et al. The calcium phosphate matrix of FGF-2-apatite composite layers contributes to their biological effects. Int J Mol Sci 2014;15:10252-70. [Crossref] [PubMed]
- Mutsuzaki H, Ito A, Sogo Y, et al. Enhanced wound healing associated with Sharpey's fiber-like tissue formation around FGF-2-apatite composite layers on percutaneous titanium screws in rabbits. Arch Orthop Trauma Surg 2012;132:113-21. [Crossref] [PubMed]
- Leedy MR, Jennings JA, Haggard WO, et al. Effects of VEGF-loaded chitosan coatings. J Biomed Mater Res A 2014;102:752-9. [Crossref] [PubMed]
- Jin K, Li B, Lou L, et al. In vivo vascularization of MSC-loaded porous hydroxyapatite constructs coated with VEGF-functionalized collagen/heparin multilayers. Sci Rep 2016;6:19871. [Crossref] [PubMed]
- Zavan B, Ferroni L, Gardin C, et al. Release of VEGF from Dental Implant Improves Osteogenetic Process: Preliminary In Vitro Tests. Materials (Basel) 2017;10:1052. [Crossref] [PubMed]
- Li Y, Liu Y, Bai H, et al. Sustained Release of VEGF to Promote Angiogenesis and Osteointegration of Three-Dimensional Printed Biomimetic Titanium Alloy Implants. Front Bioeng Biotechnol 2021;9:757767. [Crossref] [PubMed]
- Chen X, Hirt H, Li Y, et al. Antimicrobial GL13K peptide coatings killed and ruptured the wall of Streptococcus gordonii and prevented formation and growth of biofilms. PLoS One 2014;9:e111579. [Crossref] [PubMed]
- Liu J, Tang Y, Yang W, et al. Functionalization of titanium substrate with multifunctional peptide OGP-NAC for the regulation of osteoimmunology. Biomater Sci 2019;7:1463-76. [Crossref] [PubMed]
- Yakufu M, Wang Z, Wang Y, et al. Covalently functionalized poly(etheretherketone) implants with osteogenic growth peptide (OGP) to improve osteogenesis activity. RSC Adv 2020;10:9777-85. [Crossref] [PubMed]
- Liu J, Yang W, Tao B, et al. Preparing and immobilizing antimicrobial osteogenic growth peptide on titanium substrate surface. J Biomed Mater Res A 2018;106:3021-33. [Crossref] [PubMed]
- Chen D, Zhao M, Mundy GR. Bone morphogenetic proteins. Growth Factors 2004;22:233-41. [Crossref] [PubMed]
- Ebara S, Nakayama K. Mechanism for the action of bone morphogenetic proteins and regulation of their activity. Spine (Phila Pa 1976) 2002;27:S10-5. [Crossref] [PubMed]
- Haimov H, Yosupov N, Pinchasov G, et al. Bone Morphogenetic Protein Coating on Titanium Implant Surface: a Systematic Review. J Oral Maxillofac Res 2017;8:e1. [Crossref] [PubMed]
- Bergeron E, Senta H, Mailloux A, et al. Murine preosteoblast differentiation induced by a peptide derived from bone morphogenetic proteins-9. Tissue Eng Part A 2009;15:3341-9. [Crossref] [PubMed]
- Lauzon MA, Daviau A, Drevelle O, et al. Identification of a growth factor mimicking the synergistic effect of fetal bovine serum on BMP-9 cell response. Tissue Eng Part A 2014;20:2524-35. [Crossref] [PubMed]
- Mantripragada VP, Jayasuriya AC. Bone regeneration using injectable BMP-7 loaded chitosan microparticles in rat femoral defect. Mater Sci Eng C Mater Biol Appl 2016;63:596-608. [Crossref] [PubMed]
- Benington L, Rajan G, Locher C, et al. Fibroblast Growth Factor 2-A Review of Stabilisation Approaches for Clinical Applications. Pharmaceutics 2020;12:508. [Crossref] [PubMed]
- Li J, Li Z, Wang C, et al. The Regulatory Effect of VEGF-Ax on Rat Bone Marrow Mesenchymal Stem Cells' Angioblastic Differentiation and Its Proangiogenic Ability. Stem Cells Dev 2020;29:667-77. [Crossref] [PubMed]
- Kauffmann P, Raschke D, Tröltzsch M, et al. The use of rhBMP2 for augmentation of established horizontal/vertical defects may require additional use of rhVEGF to achieve significant bone regeneration: An in vivo experimental study. Clin Oral Implants Res 2021;32:1228-40. [Crossref] [PubMed]
- Kim H, Choi SH, Chung SM, et al. Enhanced bone forming ability of SLA-treated Ti coated with a calcium phosphate thin film formed by e-beam evaporation. Biomed Mater 2010;5:044106. [Crossref] [PubMed]
- Chen C, Li H, Kong X, et al. Immobilizing osteogenic growth peptide with and without fibronectin on a titanium surface: effects of loading methods on mesenchymal stem cell differentiation. Int J Nanomedicine 2015;10:283-95. [PubMed]
- Anderson JM, Patel JD. Biomaterial-Dependent Characteristics of the Foreign Body Response and S. epidermidis Biofilm Interactions. In: Moriarty TF, Zaat SAJ, Busscher HJ. editors. Biomaterials Associated Infection: Immunological Aspects and Antimicrobial Strategies. New York: Springer; 2013:119-49.
- Hancock RE, Sahl HG. Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nat Biotechnol 2006;24:1551-7. [Crossref] [PubMed]
- Dutta D, Kumar N, D P, Willcox M. Antimicrobial activity of four cationic peptides immobilised to poly-hydroxyethylmethacrylate. Biofouling 2016;32:429-38. [Crossref] [PubMed]
- Nie B, Ao H, Zhou J, et al. Biofunctionalization of titanium with bacitracin immobilization shows potential for anti-bacteria, osteogenesis and reduction of macrophage inflammation. Colloids Surf B Biointerfaces 2016;145:728-39. [Crossref] [PubMed]
- Li T, Wang N, Chen S, et al. Antibacterial activity and cytocompatibility of an implant coating consisting of TiO2 nanotubes combined with a GL13K antimicrobial peptide. Int J Nanomedicine 2017;12:2995-3007. [Crossref] [PubMed]
- Godoy-Gallardo M, Mas-Moruno C, Yu K, et al. Antibacterial properties of hLf1-11 peptide onto titanium surfaces: a comparison study between silanization and surface initiated polymerization. Biomacromolecules 2015;16:483-96. [Crossref] [PubMed]
- Geng H, Yuan Y, Adayi A, et al. Engineered chimeric peptides with antimicrobial and titanium-binding functions to inhibit biofilm formation on Ti implants. Mater Sci Eng C Mater Biol Appl 2018;82:141-54. [Crossref] [PubMed]
- Liu Z, Ma S, Duan S, et al. Modification of Titanium Substrates with Chimeric Peptides Comprising Antimicrobial and Titanium-Binding Motifs Connected by Linkers To Inhibit Biofilm Formation. ACS Appl Mater Interfaces 2016;8:5124-36. [Crossref] [PubMed]
- Yucesoy DT, Hnilova M, Boone K, et al. Chimeric peptides as implant functionalization agents for titanium alloy implants with antimicrobial properties. JOM (1989) 2015;67:754-66. [Crossref] [PubMed]
- Tan XW, Goh TW, Saraswathi P, et al. Effectiveness of antimicrobial peptide immobilization for preventing perioperative cornea implant-associated bacterial infection. Antimicrob Agents Chemother 2014;58:5229-38. [Crossref] [PubMed]
- Chen R, Willcox MD, Ho KK, et al. Antimicrobial peptide melimine coating for titanium and its in vivo antibacterial activity in rodent subcutaneous infection models. Biomaterials 2016;85:142-51. [Crossref] [PubMed]
- De Zoysa GH, Sarojini V. Feasibility Study Exploring the Potential of Novel Battacin Lipopeptides as Antimicrobial Coatings. ACS Appl Mater Interfaces 2017;9:1373-83. [Crossref] [PubMed]
- Gao G, Lange D, Hilpert K, et al. The biocompatibility and biofilm resistance of implant coatings based on hydrophilic polymer brushes conjugated with antimicrobial peptides. Biomaterials 2011;32:3899-909. [Crossref] [PubMed]
- Zhao G, Zhong H, Zhang M, et al. Effects of antimicrobial peptides on Staphylococcus aureus growth and biofilm formation in vitro following isolation from implant-associated infections. Int J Clin Exp Med 2015;8:1546-51. [PubMed]
- de Breij A, Riool M, Kwakman PH, et al. Prevention of Staphylococcus aureus biomaterial-associated infections using a polymer-lipid coating containing the antimicrobial peptide OP-145. J Control Release 2016;222:1-8. [Crossref] [PubMed]
- Mateescu M, Baixe S, Garnier T, et al. Antibacterial Peptide-Based Gel for Prevention of Medical Implanted-Device Infection. PLoS One 2015;10:e0145143. [Crossref] [PubMed]
- Cheng H, Yue K, Kazemzadeh-Narbat M, et al. Mussel-Inspired Multifunctional Hydrogel Coating for Prevention of Infections and Enhanced Osteogenesis. ACS Appl Mater Interfaces 2017;9:11428-39. [Crossref] [PubMed]
- Riool M, de Breij A, de Boer L, et al. Controlled Release of LL-37-Derived Synthetic Antimicrobial and Anti-Biofilm Peptides SAAP-145 and SAAP-276 Prevents Experimental Biomaterial-Associated Staphylococcus aureus Infection. Adv Funct Mater 2017;27:1606623. [Crossref]
- Pfeufer NY, Hofmann-Peiker K, Mühle M, et al. Bioactive coating of titanium surfaces with recombinant human β-defensin-2 (rHuβD2) may prevent bacterial colonization in orthopaedic surgery. J Bone Joint Surg Am 2011;93:840-6. [Crossref] [PubMed]
- Warnke PH, Voss E, Russo PA, et al. Antimicrobial peptide coating of dental implants: biocompatibility assessment of recombinant human beta defensin-2 for human cells. Int J Oral Maxillofac Implants 2013;28:982-8. [Crossref] [PubMed]
- Townsend L, Williams RL, Anuforom O, et al. Antimicrobial peptide coatings for hydroxyapatite: electrostatic and covalent attachment of antimicrobial peptides to surfaces. J R Soc Interface 2017;14:20160657. [Crossref] [PubMed]
- Shen X, Al-Baadani MA, He H, et al. Antibacterial and osteogenesis performances of LL37-loaded titania nanopores in vitro and in vivo. Int J Nanomedicine 2019;14:3043-54. [Crossref] [PubMed]
- He Y, Mu C, Shen X, et al. Peptide LL-37 coating on micro-structured titanium implants to facilitate bone formation in vivo via mesenchymal stem cell recruitment. Acta Biomater 2018;80:412-24. [Crossref] [PubMed]
- Makihira S, Nikawa H, Shuto T, et al. Evaluation of trabecular bone formation in a canine model surrounding a dental implant fixture immobilized with an antimicrobial peptide derived from histatin. J Mater Sci Mater Med 2011;22:2765-72. [Crossref] [PubMed]
- van Dijk IA, Beker AF, Jellema W, et al. Histatin 1 Enhances Cell Adhesion to Titanium in an Implant Integration Model. J Dent Res 2017;96:430-6. [Crossref] [PubMed]
- Sun W, Ma D, Bolscher JGM, et al. Human Salivary Histatin-1 Promotes Osteogenic Cell Spreading on Both Bio-Inert Substrates and Titanium SLA Surfaces. Front Bioeng Biotechnol 2020;8:584410. [Crossref] [PubMed]
- Siwakul P, Sirinnaphakorn L, Suwanprateep J, et al. Cellular responses of histatin-derived peptides immobilized titanium surface using a tresyl chloride-activated method. Dent Mater J 2021;40:934-41. [Crossref] [PubMed]
- Pazgier M, Hoover DM, Yang D, et al. Human beta-defensins. Cell Mol Life Sci 2006;63:1294-313. [Crossref] [PubMed]
- Abiko Y, Nishimura M, Kusano K, et al. Upregulated expression of human beta defensin-1 and -3 mRNA during differentiation of keratinocyte immortalized cell lines, HaCaT and PHK16-0b. J Dermatol Sci 2003;31:225-8. [Crossref] [PubMed]
- Donald CD, Sun CQ, Lim SD, et al. Cancer-specific loss of beta-defensin 1 in renal and prostatic carcinomas. Lab Invest 2003;83:501-5. [Crossref] [PubMed]
- Varoga D, Pufe T, Harder J, et al. Human beta-defensin 3 mediates tissue remodeling processes in articular cartilage by increasing levels of metalloproteinases and reducing levels of their endogenous inhibitors. Arthritis Rheum 2005;52:1736-45. [Crossref] [PubMed]
- Schneider JJ, Unholzer A, Schaller M, et al. Human defensins. J Mol Med (Berl) 2005;83:587-95. [Crossref] [PubMed]
- Schröder JM, Harder J. Human beta-defensin-2. Int J Biochem Cell Biol 1999;31:645-51. [Crossref] [PubMed]
- Zanetti M. Cathelicidins, multifunctional peptides of the innate immunity. J Leukoc Biol 2004;75:39-48. [Crossref] [PubMed]
- Puri S, Edgerton M. How does it kill?: understanding the candidacidal mechanism of salivary histatin 5. Eukaryot Cell 2014;13:958-64. [Crossref] [PubMed]
- Saha S, Pramanik K, Biswas A. Silk fibroin coated TiO(2) nanotubes for improved osteogenic property of Ti6Al4V bone implants. Mater Sci Eng C Mater Biol Appl 2019;105:109982. [Crossref] [PubMed]
- Chen X, Gao Y, Wang Y, et al. Mussel-inspired peptide mimicking: An emerging strategy for surface bioengineering of medical implants. Smart Mater Med 2021;2:26-37. [Crossref]
- Dinerman AA, Cappello J, Ghandehari H, et al. Swelling behavior of a genetically engineered silk-elastinlike protein polymer hydrogel. Biomaterials 2002;23:4203-10. [Crossref] [PubMed]
- Das S, Pati F, Choi YJ, et al. Bioprintable, cell-laden silk fibroin-gelatin hydrogel supporting multilineage differentiation of stem cells for fabrication of three-dimensional tissue constructs. Acta Biomater 2015;11:233-46. [Crossref] [PubMed]
- Li C, Tong L. Topology optimization of incompressible materials based on the mixed SBFEM. Computers & Structures 2016;165:24-33. [Crossref]
- Wenhao Z, Zhang T, Yan J, et al. In vitro and in vivo evaluation of structurally-controlled silk fibroin coatings for orthopedic infection and in-situ osteogenesis. Acta Biomater 2020;116:223-45. [Crossref] [PubMed]
- Naskar D, Nayak S, Dey T, et al. Non-mulberry silk fibroin influence osteogenesis and osteoblast-macrophage cross talk on titanium based surface. Sci Rep 2014;4:4745. [Crossref] [PubMed]
- Lovati AB, Lopa S, Bottagisio M, et al. Peptide-Enriched Silk Fibroin Sponge and Trabecular Titanium Composites to Enhance Bone Ingrowth of Prosthetic Implants in an Ovine Model of Bone Gaps. Front Bioeng Biotechnol 2020;8:563203. [Crossref] [PubMed]
- Asadi H, Ghalei S, Handa H, et al. Cellulose nanocrystal reinforced silk fibroin coating for enhanced corrosion protection and biocompatibility of Mg-based alloys for orthopedic implant applications. Prog Org Coat 2021;161:106525. [Crossref]
- Brubaker CE, Messersmith PB. The present and future of biologically inspired adhesive interfaces and materials. Langmuir 2012;28:2200-5. [Crossref] [PubMed]
- Waite JH, Tanzer ML. Polyphenolic Substance of Mytilus edulis: Novel Adhesive Containing L-Dopa and Hydroxyproline. Science 1981;212:1038-40. [Crossref] [PubMed]
- Lee H, Dellatore SM, Miller WM, et al. Mussel-inspired surface chemistry for multifunctional coatings. Science 2007;318:426-30. [Crossref] [PubMed]
- Sun J, Huang Y, Zhao H, et al. Bio-clickable mussel-inspired peptides improve titanium-based material osseointegration synergistically with immunopolarization-regulation. Bioact Mater 2021;9:1-14. [Crossref] [PubMed]
- Zhao H, Wang X, Zhang W, et al. Bioclickable Mussel-Derived Peptides With Immunoregulation for Osseointegration of PEEK. Front Bioeng Biotechnol 2021;9:780609. [Crossref] [PubMed]
- Wang H, Lin C, Zhang X, et al. Mussel-Inspired Polydopamine Coating: A General Strategy To Enhance Osteogenic Differentiation and Osseointegration for Diverse Implants. ACS Appl Mater Interfaces 2019;11:7615-25. [Crossref] [PubMed]
- Yazici H, Fong H, Wilson B, et al. Biological response on a titanium implant-grade surface functionalized with modular peptides. Acta Biomater 2013;9:5341-52. [Crossref] [PubMed]
- Bierbaum S, Hempel U, Geissler U, et al. Modification of Ti6AL4V surfaces using collagen I, III, and fibronectin. II. Influence on osteoblast responses. J Biomed Mater Res A 2003;67:431-8. [Crossref] [PubMed]
- Bierbaum S, Beutner R, Hanke T, et al. Modification of Ti6Al4V surfaces using collagen I, III, and fibronectin. I. Biochemical and morphological characteristics of the adsorbed matrix. J Biomed Mater Res A 2003;67:421-30. [Crossref] [PubMed]
- Ku Y, Chung CP, Jang JH. The effect of the surface modification of titanium using a recombinant fragment of fibronectin and vitronectin on cell behavior. Biomaterials 2005;26:5153-7. [Crossref] [PubMed]
- Wisdom EC, Zhou Y, Chen C, et al. Mitigation of peri-implantitis by rational design of bifunctional peptides with antimicrobial properties. ACS Biomater Sci Eng 2020;6:2682-95. [Crossref] [PubMed]
- Raoufinia R, Mota A, Keyhanvar N, et al. Overview of Albumin and Its Purification Methods. Adv Pharm Bull 2016;6:495-507. [Crossref] [PubMed]
- Höhn S, Braem A, Neirinck B, et al. Albumin coatings by alternating current electrophoretic deposition for improving corrosion resistance and bioactivity of titanium implants. Mater Sci Eng C Mater Biol Appl 2017;73:798-807. [Crossref] [PubMed]
- Sivaraman B, Latour RA. The relationship between platelet adhesion on surfaces and the structure versus the amount of adsorbed fibrinogen. Biomaterials 2010;31:832-9. [Crossref] [PubMed]
- Hylton DM, Shalaby SW, Latour RA Jr. Direct correlation between adsorption-induced changes in protein structure and platelet adhesion. J Biomed Mater Res A 2005;73:349-58. [Crossref] [PubMed]
- Peters T Jr. All about albumin: biochemistry, genetics, and medical applications. Cambridge: Academic Press; 1995.
- Feng J, Wu G, Qing C. Biomimetic synthesis of hollow calcium carbonate with the existence of the agar matrix and bovine serum albumin. Mater Sci Eng C Mater Biol Appl 2016;58:409-11. [Crossref] [PubMed]
- Khan MA, Williams RL, Williams DF. Conjoint corrosion and wear in titanium alloys. Biomaterials 1999;20:765-72. [Crossref] [PubMed]
- Kinnari TJ, Peltonen LI, Kuusela P, et al. Bacterial adherence to titanium surface coated with human serum albumin. Otol Neurotol 2005;26:380-4. [Crossref] [PubMed]
- Zhang Y, Addison O, Yu F, et al. Time-dependent Enhanced Corrosion of Ti6Al4V in the Presence of H(2)O(2) and Albumin. Sci Rep 2018;8:3185. [Crossref] [PubMed]
- Eckrich J, Hoormann N, Kersten E, et al. Surface Modification of Porous Polyethylene Implants with an Albumin-Based Nanocarrier-Release System. Biomedicines 2021;9:1485. [Crossref] [PubMed]
- Duan P, Bonewald LF. The role of the wnt/β-catenin signaling pathway in formation and maintenance of bone and teeth. Int J Biochem Cell Biol 2016;77:23-9. [Crossref] [PubMed]
- Florio M, Gunasekaran K, Stolina M, et al. A bispecific antibody targeting sclerostin and DKK-1 promotes bone mass accrual and fracture repair. Nat Commun 2016;7:11505. [Crossref] [PubMed]
- Korn P, Kramer I, Schlottig F, et al. Systemic sclerostin antibody treatment increases osseointegration and biomechanical competence of zoledronic-acid-coated dental implants in a rat osteoporosis model. Eur Cell Mater 2019;37:333-46. [Crossref] [PubMed]
- Delgado-Calle J, Sato AY, Bellido T. Role and mechanism of action of sclerostin in bone. Bone 2017;96:29-37. [Crossref] [PubMed]
- Baron R, Kneissel M. WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat Med 2013;19:179-92. [Crossref] [PubMed]
- Leupin O, Piters E, Halleux C, et al. Bone overgrowth-associated mutations in the LRP4 gene impair sclerostin facilitator function. J Biol Chem 2011;286:19489-500. [Crossref] [PubMed]
- Yu SH, Hao J, Fretwurst T, et al. Sclerostin-Neutralizing Antibody Enhances Bone Regeneration Around Oral Implants. Tissue Eng Part A 2018;24:1672-9. [Crossref] [PubMed]
- Yao Y, Kauffmann F, Maekawa S, et al. Sclerostin antibody stimulates periodontal regeneration in large alveolar bone defects. Sci Rep 2020;10:16217. [Crossref] [PubMed]
- Han XL, Liu M, Voisey A, et al. Post-natal effect of overexpressed DKK1 on mandibular molar formation. J Dent Res 2011;90:1312-7. [Crossref] [PubMed]
- Napimoga MH, Nametala C, da Silva FL, et al. Involvement of the Wnt-β-catenin signalling antagonists, sclerostin and dickkopf-related protein 1, in chronic periodontitis. J Clin Periodontol 2014;41:550-7. [Crossref] [PubMed]
- Boyden LM, Mao J, Belsky J, et al. High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med 2002;346:1513-21. [Crossref] [PubMed]
- Liu M, Kurimoto P, Zhang J, et al. Sclerostin and DKK1 Inhibition Preserves and Augments Alveolar Bone Volume and Architecture in Rats with Alveolar Bone Loss. J Dent Res 2018;97:1031-8. [Crossref] [PubMed]
- Leung EKY. Parathyroid hormone. Adv Clin Chem 2021;101:41-93. [Crossref] [PubMed]
- Silva BC, Bilezikian JP. Parathyroid hormone: anabolic and catabolic actions on the skeleton. Curr Opin Pharmacol 2015;22:41-50. [Crossref] [PubMed]
- Dobnig H, Turner RT. The effects of programmed administration of human parathyroid hormone fragment (1-34) on bone histomorphometry and serum chemistry in rats. Endocrinology 1997;138:4607-12. [Crossref] [PubMed]
- Lai K, Xi Y, Miao X, et al. PTH coatings on titanium surfaces improved osteogenic integration by increasing expression levels of BMP-2/Runx2/Osterix. RSC Adv 2017;7:56256-65. [Crossref]
- Canalis E, Giustina A, Bilezikian JP. Mechanisms of anabolic therapies for osteoporosis. N Engl J Med 2007;357:905-16. [Crossref] [PubMed]
- Neuprez A, Reginster JY. Bone-forming agents in the management of osteoporosis. Best Pract Res Clin Endocrinol Metab 2008;22:869-83. [Crossref] [PubMed]
- Ardura JA, Portal-Núñez S, Lozano D, et al. Local delivery of parathyroid hormone-related protein-derived peptides coated onto a hydroxyapatite-based implant enhances bone regeneration in old and diabetic rats. J Biomed Mater Res A 2016;104:2060-70. [Crossref] [PubMed]
- Tang J, Yan D, Chen L, et al. Enhancement of local bone formation on titanium implants in osteoporotic rats by biomimetic multilayered structures containing parathyroid hormone (PTH)-related protein. Biomed Mater 2020;15:045011. [Crossref] [PubMed]
- Fang W, Zhao S, He F, et al. Influence of simvastatin-loaded implants on osseointegration in an ovariectomized animal model. Biomed Res Int 2015;2015:831504. [Crossref] [PubMed]
- Javed F, Al Amri MD, Kellesarian SV, et al. Efficacy of parathyroid hormone supplementation on the osseointegration of implants: a systematic review. Clin Oral Investig 2016;20:649-58. [Crossref] [PubMed]
- Wang C, Wang S, Yang Y, et al. Bioinspired, biocompatible and peptide-decorated silk fibroin coatings for enhanced osteogenesis of bioinert implant. J Biomater Sci Polym Ed 2018;29:1595-611. [Crossref] [PubMed]
- Calvo-Guirado JL, Mate-Sanchez J, Delgado-Ruiz R, et al. Effects of growth hormone on initial bone formation around dental implants: a dog study. Clin Oral Implants Res 2011;22:587-93. [Crossref] [PubMed]
- Muñoz F, López-Peña M, Miño N, et al. Topical application of melatonin and growth hormone accelerates bone healing around dental implants in dogs. Clin Implant Dent Relat Res 2012;14:226-35. [Crossref] [PubMed]
- Abreu ME, Valiati R, Hubler R, et al. Effect of Recombinant Human Growth Hormone on Osseointegration of Titanium Implants: A Histologic and Biomechanical Study in Rabbits. J Oral Implantol 2015;41:e102-9. [Crossref] [PubMed]
- Salomó-Coll O, Maté-Sánchez de Val JE, Ramírez-Fernández MP, et al. Osseoinductive elements for promoting osseointegration around immediate implants: a pilot study in the foxhound dog. Clin Oral Implants Res 2016;27:e167-75. [Crossref] [PubMed]
- Sun T, Li J, Xing HL, et al. Melatonin improves the osseointegration of hydroxyapatite-coated titanium implants in senile female rats. Z Gerontol Geriatr 2020;53:770-7. [Crossref] [PubMed]
- Cerqueira A, Romero-Gavilán F, Araújo-Gomes N, et al. A possible use of melatonin in the dental field: Protein adsorption and in vitro cell response on coated titanium. Mater Sci Eng C Mater Biol Appl 2020;116:111262. [Crossref] [PubMed]
- Nabeshima A, Pajarinen J, Lin TH, et al. Mutant CCL2 protein coating mitigates wear particle-induced bone loss in a murine continuous polyethylene infusion model. Biomaterials 2017;117:1-9. [Crossref] [PubMed]
- Keeney M, Waters H, Barcay K, et al. Mutant MCP-1 protein delivery from layer-by-layer coatings on orthopedic implants to modulate inflammatory response. Biomaterials 2013;34:10287-95. [Crossref] [PubMed]
- Sato T, Pajarinen J, Behn A, et al. The effect of local IL-4 delivery or CCL2 blockade on implant fixation and bone structural properties in a mouse model of wear particle induced osteolysis. J Biomed Mater Res A 2016;104:2255-62. [Crossref] [PubMed]
- Abduljabbar T, Kellesarian SV, Vohra F, et al. Effect of Growth Hormone Supplementation on Osseointegration: A Systematic Review and Meta-analyses. Implant Dent 2017;26:613-20. [Crossref] [PubMed]
- Klein IE. The effect of thyrocalcitonin and growth hormones on bone metabolism. J Prosthet Dent 1975;33:365-79. [Crossref] [PubMed]
- Ohlsson C, Vidal O. Effects of growth hormone and insulin-like growth factors on human osteoblasts. Eur J Clin Invest 1998;28:184-6. [Crossref] [PubMed]
- Kim Y, Hong JW, Chung YS, et al. Efficacy and safety of sustained-release recombinant human growth hormone in Korean adults with growth hormone deficiency. Yonsei Med J 2014;55:1042-8. [Crossref] [PubMed]
- Guicheux J, Gauthier O, Aguado E, et al. Human growth hormone locally released in bone sites by calcium-phosphate biomaterial stimulates ceramic bone substitution without systemic effects: a rabbit study. J Bone Miner Res 1998;13:739-48. [Crossref] [PubMed]
- Nakade O, Koyama H, Ariji H, et al. Melatonin stimulates proliferation and type I collagen synthesis in human bone cells in vitro. J Pineal Res 1999;27:106-10. [Crossref] [PubMed]
- Johannsson G, Rosén T, Bosaeus I, et al. Two years of growth hormone (GH) treatment increases bone mineral content and density in hypopituitary patients with adult-onset GH deficiency. J Clin Endocrinol Metab 1996;81:2865-73. [PubMed]
- Hansen TB, Brixen K, Vahl N, et al. Effects of 12 months of growth hormone (GH) treatment on calciotropic hormones, calcium homeostasis, and bone metabolism in adults with acquired GH deficiency: a double blind, randomized, placebo-controlled study. J Clin Endocrinol Metab 1996;81:3352-9. [PubMed]
- Permuy M, López-Peña M, González-Cantalapiedra A, et al. Melatonin: A Review of Its Potential Functions and Effects on Dental Diseases. Int J Mol Sci 2017;18:865. [Crossref] [PubMed]
- Redman J, Armstrong S, Ng KT. Free-running activity rhythms in the rat: entrainment by melatonin. Science 1983;219:1089-91. [Crossref] [PubMed]
- Dollins AB, Zhdanova IV, Wurtman RJ, et al. Effect of inducing nocturnal serum melatonin concentrations in daytime on sleep, mood, body temperature, and performance. Proc Natl Acad Sci U S A 1994;91:1824-8. [Crossref] [PubMed]
- García-Mauriño S, Pozo D, Calvo JR, et al. Correlation between nuclear melatonin receptor expression and enhanced cytokine production in human lymphocytic and monocytic cell lines. J Pineal Res 2000;29:129-37. [Crossref] [PubMed]
- Gibon E, Ma T, Ren PG, et al. Selective inhibition of the MCP-1-CCR2 ligand-receptor axis decreases systemic trafficking of macrophages in the presence of UHMWPE particles. J Orthop Res 2012;30:547-53. [Crossref] [PubMed]
- Gong JH, Clark-Lewis I. Antagonists of monocyte chemoattractant protein 1 identified by modification of functionally critical NH2-terminal residues. J Exp Med 1995;181:631-40. [Crossref] [PubMed]
- Yao Z, Keeney M, Lin TH, et al. Mutant monocyte chemoattractant protein 1 protein attenuates migration of and inflammatory cytokine release by macrophages exposed to orthopedic implant wear particles. J Biomed Mater Res A 2014;102:3291-7. [Crossref] [PubMed]
- Jiang X, Sato T, Yao Z, et al. Local delivery of mutant CCL2 protein-reduced orthopaedic implant wear particle-induced osteolysis and inflammation in vivo. J Orthop Res 2016;34:58-64. [Crossref] [PubMed]
- James AW, LaChaud G, Shen J, et al. A Review of the Clinical Side Effects of Bone Morphogenetic Protein-2. Tissue Eng Part B Rev 2016;22:284-97. [Crossref] [PubMed]
- Vidya M, Rajagopal S. Silk Fibroin: A Promising Tool for Wound Healing and Skin Regeneration. Int J Polym Sci 2021;2021:9069924. [Crossref]
- Aditya S, Rattan A. Sclerostin Inhibition: A Novel Target for the Treatment of Postmenopausal Osteoporosis. J Midlife Health 2021;12:267-75. [Crossref] [PubMed]
Cite this article as: De Leon J, Romanos GE. The use of bioactive proteins and peptides to promote osseointegration and success of dental implants: a narrative review. Front Oral Maxillofac Med 2025;7:18.