Next Article in Journal
Fully Automatic Liver and Tumor Segmentation from CT Image Using an AIM-Unet
Previous Article in Journal
Preparation and Characterization of Biomimetic Functional Scaffold with Gradient Structure for Osteochondral Defect Repair
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Peptides in Dentistry: A Scoping Review

by
Louis Hardan
1,†,
Jean Claude Abou Chedid
2,†,
Rim Bourgi
1,3,
Carlos Enrique Cuevas-Suárez
4,*,
Monika Lukomska-Szymanska
5,
Vincenzo Tosco
6,
Ana Josefina Monjarás-Ávila
4,
Massa Jabra
7,
Fouad Salloum-Yared
8,
Naji Kharouf
3,9,*,
Davide Mancino
3,9,10 and
Youssef Haikel
3,9,10,*
1
Department of Restorative Dentistry, School of Dentistry, Saint Joseph University, Beirut 1107 2180, Lebanon
2
Department of Pediatric Dentistry, Faculty of Dentistry, Saint Joseph University, Beirut 1107 2180, Lebanon
3
Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France
4
Dental Materials Laboratory, Academic Area of Dentistry, Autonomous University of Hidalgo State, San Agustín Tlaxiaca 42160, Mexico
5
Department of General Dentistry, Medical University of Lodz, 251 Pomorska St., 92-213 Lodz, Poland
6
Department of Clinical Sciences and Stomatology (DISCO), Polytechnic University of Marche, 60126 Ancona, Italy
7
Faculty of Medicine, Damascus University, Damascus 0100, Syria
8
Private Practice, 54290 Trier, Germany
9
Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
10
Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Bioengineering 2023, 10(2), 214; https://doi.org/10.3390/bioengineering10020214
Submission received: 18 January 2023 / Revised: 2 February 2023 / Accepted: 3 February 2023 / Published: 6 February 2023
(This article belongs to the Section Regenerative Engineering)

Abstract

:
Currently, it remains unclear which specific peptides could be appropriate for applications in different fields of dentistry. The aim of this scoping review was to scan the contemporary scientific papers related to the types, uses and applications of peptides in dentistry at the moment. Literature database searches were performed in the following databases: PubMed/MEDLINE, Scopus, Web of Science, Embase, and Scielo. A total of 133 articles involving the use of peptides in dentistry-related applications were included. The studies involved experimental designs in animals, microorganisms, or cells; clinical trials were also identified within this review. Most of the applications of peptides included caries management, implant osseointegration, guided tissue regeneration, vital pulp therapy, antimicrobial activity, enamel remineralization, periodontal therapy, the surface modification of tooth implants, and the modification of other restorative materials such as dental adhesives and denture base resins. The in vitro and in vivo studies included in this review suggested that peptides may have beneficial effects for treating early carious lesions, promoting cell adhesion, enhancing the adhesion strength of dental implants, and in tissue engineering as healthy promotors of the periodontium and antimicrobial agents. The lack of clinical trials should be highlighted, leaving a wide space available for the investigation of peptides in dentistry.

1. Introduction

Dental plaques contain over 750 different bacterial species, which are the major reason for dental caries, with streptococci being the most predominantly present. These bacteria, due to the production of acids, can demineralize and affect mineralized tooth tissues [1]. Different additives and biomaterials were used in dental treatments in order to eliminate and decrease the number of bacteria in the oral cavity and teeth tissues. Some dental materials, such as calcium silicate-based products, have been introduced in the dental market due to their antibacterial, antioxidant and remineralization properties [2]. Other solutions that have antibacterial effects are used to clean the root canal and kill resistant bacteria in the root canal system [3]. Even though they sometimes display high cytotoxicity [4,5], these materials are still currently used in dentistry.
It should be remembered that a peptide is expressed as a short polymer of amino acids (AA) [6]. According to the description of diverse authors in the literature, sizes of peptides may vary from <20, <50, to <100 [7,8,9,10]. The use of peptides has been paid attention to over the last two decades [6,11]. These peptides were used in various dental fields such as in endodontic treatment, coronal restoration, caries management, bone and dental tissue remineralization and in the modification of dental materials in order to promote the biological effects of these materials in the oral environment [6,12].
In the last periods, over 7000 native peptides (NP) have been considered by means of significant human physiological functions [13]. These peptides have functions by way of cell-penetrating, cell adhesion motifs, tumor-homing peptides, neuropeptides, structural peptides, peptide hormones, antimicrobial peptides, peptide tags, matrix metalloprotease substrates, growth factors, amyloid peptides, and erstwhile diverse NPs [10].
Nevertheless, one should state that NPs are frequently not truthfully appropriate for therapeutic usage since they have intrinsic drawbacks, including their poor physical and chemical stability, low oral bioavailability, short flowing plasma half-life, and quick removal from the circulation through the kidneys and the liver [9,13,14]. It is well described that peptides such as insulin and adrenocorticotrophic hormone were used for human therapeutic purposes in the first half of the 20th century [15]. Later on, synthetic oxytocin and vasopressin arrived in clinical use in the 1950s with the chemical elucidation of the sequences of these peptides [16].
Lately, pharmaceutical manufacturing has amplified the consideration of novel therapeutic peptides, persistently touching clinical claims [9,17]. By 2018, more than 60 peptides were approved by the Food and Drug Administration (FDA), and more than 600 were undergoing preclinical and clinical examinations [9,18]. With the current elaborations of solid-phase peptide synthesis, the production of therapeutic synthetic peptides (SP) has become achievable [9]. Accordingly, innovative synthetic approaches permit the modulation of pharmacokinetic assets and focus on specificity through AAs, the integration of non-natural AAs, backbone adjustments, and the peptide conjugates refining solubility or prolonging the half-life [8,13,14].
It is recognized that human dental masses, once fashioned, cannot be biologically replaced or repaired, and their multifaceted conformations require diverse approaches for regeneration [6]. However, it is unclear in the literature which specific peptides could be effective for applications in different fields of dentistry. Thus, the aim of this scoping review was to map the contemporary scientific papers related to the use and applications of peptides in dentistry at present.

2. Materials and Methods

The present scoping review has been described according to the PRISMA extension for scoping reviews guideline [19]. The review protocol was registered at Open Science Framework, and it is available at https://osf.io/up6ty (accessed on 18 December 2022). The systematic search was performed according to the following parameters: (i) population: peer-reviewed articles; (ii) intervention: use of natural or synthetic peptides; (iii) comparison: other substances or treatments; (iv) outcome: dental applications, (v) study design: in vitro or in vivo articles. The general question of the review was as follows: what scientific applications of products based on peptides are being used for dental applications?

2.1. Information Sources and Search

The literature database search was performed by two independent reviewers (RB and CECS) until September 2022. The search was carried out in the following databases: PubMed/MEDLINE, Scopus, Web of Science, Embase, and Scielo. The search strategy was first defined for the MEDLINE database using a controlled vocabulary and free keywords (Table 1). The MEDLINE search strategy was then adapted to other electronic databases. The reviewers also hand-searched the reference lists of the included articles to identify additional manuscripts.

2.2. Selection Process and Data Collection Process

After running the search strategy, a reference management program was used (EndNote X9, Clarivate Analytics, Philadelphia, PA, USA) to store the files of all databases. Then, duplicate articles were removed, followed by manual removal after the organization of titles in alphabetical order. All studies were initially scanned for relevance by title followed by abstract using an online software program (Rayyan, Qatar Computing Research Institute, HBKU, Doha, Qatar). The titles and abstracts of the articles were screened according to the following inclusion criterium: in vitro or in vivo studies that evaluated or reported the use of peptides for dental applications. The search was carried out on documents published in any language without restrictions on their date of publication. Reviews, case reports, case series, pilot studies, and conference abstracts were excluded. If the review authors were not sure about the eligibility of any study, it was kept for the next phase. All phases were carried out by two independent reviewers (RB and CECS) to check whether they met the inclusion criteria. The same two reviewers summarized and categorized the data using a standardized form. The information collected included the type of study, the peptide used, the application proposed and the main results.

3. Results

This scoping review is described according to the PRISMA extension for scoping reviews guideline [19]. After database screening and duplicate removal, a total of 6450 articles were recognized (Figure 1). After title and abstract screening, 156 articles remained for full-text inspection. From the 156 articles assessed for eligibility, 23 articles were excluded due to the following reasons: in 11 articles, the full text was not retrieved [20,21,22,23,24,25,26,27,28,29,30], 4 articles were not related to the dentistry field [31,32,33,34], 4 studies were reviews [6,12,35,36], 3 studies were not related to peptides [37,38,39], and 1 study was a pilot clinical trial [40]. Thus, a total of 133 articles were included in the present review.

3.1. Characteristics of Studies

The main characteristics of the studies included in the present review are presented in Table 2.
The studies included experiments in animals and/or using bacteria or cells; also, several clinical trials were found. Most of the applications of the peptides included caries management, implant osseointegration, guided tissue regeneration, vital pulp therapy, antimicrobial activity, enamel remineralization, occlusion of dentin tubules, periodontal therapy, the surface modification of dental implants, and the modification of dental materials such as dental adhesives and denture base resins.

3.2. Synthesis of Results and Summary of Evidence

The in vitro and in vivo studies included in the present review stated that peptides may have beneficial effects for treating early carious lesions. Additionally, the use of peptides seems to be beneficial for promoting cell adhesion and enhancing the adhesion strength of dental implants. In addition, peptides were useful for tissue engineering for cell-based pulp regeneration. Peptides were also successfully used as healthy promotors of the periodontium, acting as inflammatory mediators. Finally, most peptides were used as effective antimicrobial agents.

4. Discussion

A scoping review was performed regarding the use and applications of peptides in the dental field at present. Appropriately, most of the applications of the peptides included caries management, implant osseointegration, guided tissue regeneration, vital pulp therapy, antimicrobial activity, enamel remineralization, occlusion of dentin tubules, periodontal therapy, the surface modification of dental implants, and the modification of dental materials such as dental adhesives and denture base resins.
One should keep in mind that dental caries is considered the most common disease worldwide [171], and it can lead to the destruction of dental surfaces by means of acidogenic bacteria changing sugars to acids [43]. Dissolution of the mineral tooth structure begins with caries formation, therefore generating a demineralized subsurface lesion body, similar to white spots [172], followed by the development of irreversible cavitation of the mineralized surface layer [173,174]. Treatment of manifested caries involves an oral hygiene regulation and a follow-up visit to identify whether the caries has been prevented or has advanced into a cavity, which is subsequently treated by means of restoration [173]. The use of fluoride varnish can prevent caries formation by reinforcing the inorganic surface layer, consequently inhibiting the progression of caries [175,176,177]. Fluoride ions are preserved within the inorganic surface layer covering the demineralized carious lesion due to the high correspondence to hydroxyapatite [178]. Subsequently, the demineralized subsurface zone is not penetrated by fluoride; yet this is where remineralization would be essential in an attempt to regenerate decayed enamel tissue [43]. For this reason, novel methods for the treatment of caries have been introduced to mimic the structure of the enamel matrix, such as guided enamel regeneration (GER) [179].
It should be noted that self-assembling peptide (SAP) technology was designated on the reasonable design of a short hydrophilic peptide in combination with GER that builds into fibers, establishing a three-dimensional (3D) scaffold [180,181,182]. The surface features of the fibers might fluctuate, concurring with the physiological desires of the treated tissue [66,183]. This could be explained by the rational design criteria [183]. When treating early caries lesions, SAP P11-4 fibers have been adjusted to suitably bind ionic calcium and template hydroxyapatite formation, thus, accompanying remineralization in a comparable approach of amelogenin that supports the construction of the enamel. From this analysis, the SAP P11-4 fibers might be known as a biomimetic agent [66,74]. This could be in agreement with the finding of this review that demonstrated the potential effect of peptide P11-4 in caries management.
With regards to implant osseointegration, pure titanium is commercially used for implants in the dental field due to its possible resistance to corrosion, biocompatibility, and suitable mechanical properties [184,185,186]. Researchers have detected peri-implant bone resorption produced by peri-implantitis, which is considered the key reason for the failure of osseointegrated dental implants [187,188]. In this manner, surface modification of dental implants has been a topic of interest for researchers since titanium is an inert material that decreases the aptitude for remedial tissue therapy to succeed and resists bacterial settlement [189,190,191]. To counteract peri-implantitis and advance osseointegration, different type of coatings have been investigated [192]. Surfaces incorporating chlorhexidine, antimicrobial agents and antibiotics such as gentamicin, and surfaces incorporating chlorhexidine, poly-lysine, sliver, and chitosan have all been established for coating the titanium surface of implants [52]. However, some drawbacks could be noted with antibiotic-coated titanium, such as the controversial opinion about their bacterial resistance and host cytotoxicity [193]. In 2015, Zhou et al. demonstrated that antimicrobial peptides provided a promising bifunctional titanium surface and enhanced its bactericidal activity and cytocompatibility [168]. Likewise, a previous report suggested that after 6 weeks of implantation in rabbit femurs, titanium dental implants with an antimicrobial peptide GL13K coating allowed in vivo dental implant osseointegration at similar bone growth rates to gold-standard non-coated dental implants [52]. This could be explained by the fact that GL13K is bactericidal in solution against Escherichia coli, Pseudomonas aeruginosa, Porphyromonas gingivalis and Streptococcus gordonii [83,194,195]. Similarly, Yoshinari et al. proved that the antimicrobial and titanium-binding peptides were favorable for the diminution of biofilm formation on titanium surfaces [162]. In addition, a laminin-derived peptide was demonstrated to improve and enhance the integration of soft tissue on dental titanium implants [143]. Furthermore, an epithelial basement membrane was formed on a titanium surface when platelet-activating peptide was used [135]. All in all, this could clearly support the result of this review that the use of peptides seems to be beneficial for promoting cell adhesion and enhancing the adhesion strength of dental implants.
In addition, this analysis determined that peptides were useful for guided tissue regeneration [42]. This could be achieved when a combination of a synthetic peptide named P-15 (analog of collagen) and an anorganic bovine bone mineral (ABM) was used. ABM enhanced cell attachment by differentiation and cell binding, thus enhancing osseous formation and ensuing an accelerated periodontal ligament fibroblast attachment [109,196]. Adding to P-15, biocompatible and osteoconductive filler material was thus detected [42].
A major task in the use of tissue engineering for therapy in dentistry involves the initiation of tooth and bone regeneration. The dentin phosphophoryn-derived arginine-glycine-aspartic acid-containing peptide was demonstrated as a biodegradable, biocompatible, and bioactive material for dentin regeneration. These results could be clarified by the short AA sequences of the peptide used and by its 3D conformation essential for acquiring this function [46]. Accordingly, the peptide can be used in vital pulp therapy when a specific sequence is used.
Further, most peptides were used as effective antimicrobial agents. Peptide hydrogels have shown that ultrashort peptides (<8 amino acids) might self-assemble into hydrogels. These ultrashort peptides might be intended to integrate antimicrobial motifs, such as positively charged lysine residues; thus, the peptides have integral antimicrobial features [47]. The scheme and synthesis of biocompatible hydrogels with antimicrobial activity are of numerous interests for tissue engineering drives comprising the replacement of tissue in infected root canals [65,197,198]. Moreover, antimicrobial peptides were used in coated titanium surfaces [168], dental adhesives [147], caries infection [102], and plaque biofilm inhibition [36].
Peptides were also successful for enamel remineralization. It is imperative to note that the acidic nature of dental cavities created by a massive amount of sugar intake leads to bacterial colonization and a reduction in the pH. Accordingly, the demineralization of the enamel surface begins [48]. In order to prevent this issue, numerous remineralizing agents were presented [48]. A perfect agent should be free of toxicity and qualified to initiate remineralization without any harm to the dental surface. Matrix-facilitated mineralization equal to a natural process should be carried out, though this ability is absent in almost all these agents [199]. The arrival of SAP P11-4 has overwhelmed this restriction. It has the ability to regenerate enamel. In addition, these agents initiate remineralization by making 3D constructions mimic the extracellular matrix of the dental surface [200]. Therefore, when talking about enamel remineralization, clinicians should focus on SAP due to its efficient and effective outcomes obtained in this review.
The occlusion of dentin tubules is considered possible with the help of peptides. This theory became conceivable when mineral particles were observed on dentinal tubules, thus reducing dentinal permeability and enhancing the seal of the material-tooth interfaces [57]. Bonding agents and desensitizers have been demonstrated to be effective for occluding tubules by mineral precipitation; however, these techniques are sensitive, and the long-term performance of the resin is doubtful [201,202]. As a balancing method for the protein mediation of hydroxyapatite mineralization, streamlined synthetic cationic macromolecules comprising poly(L-lysine) (PLL) that cover primary and secondary amine groups are organizationally comparable to the functional areas of the natural proteins and have further been presented to encourage silicification [203]. This review implies that this peptide-catalyst-mediated method of mineral formation for occluding tubules and/or reinforcing dentin-bonding resins might retain function on the dentin surface, advising a wide range of protective and treatment plans.
Peptides have also been successfully used as healthy promotors of the periodontium, acting as inflammatory mediators. Periodontitis is a chronic inflammatory and tissue-destructive illness. Meanwhile, the oral cavity with its polymicrobial effect makes it problematic to treat; thus, new healing approaches are mandatory. In a minimally invasive way, SAP delivers the benefit of being functional at a defect site without creating a toxic area [204]. Furthermore, their tunable mechanical characteristics and reasonably designed physicochemical features permit a high variety of encapsulated drugs [205]. Some peptides called P11-4 and P11-28/29 were considered SAP-applicable for periodontal therapy, due to their biocompatibility, injectability, tunable mechanical and physicochemical properties, and cargo-loading capacity [72].
Finally, peptides were used in the modification of dental materials such as dental adhesives and denture base resins. Recurrent decay that grows at the composite-tooth interface was demonstrated to be a disadvantage when using resin-based composite [163]. Primarily, the composite-tooth interface becomes coated by a low-viscosity adhesive system; however, when a fragile seal to the dentin is obtained, damage from enzymes, acids, and oral fluids will be achieved. This impairment is chief in crevices that are occupied by cariogenic bacteria such as Streptococcus mutans [206,207,208,209]. Various bacterial-inhibition strategies have been incorporated into adhesive systems, but none of these strategies address the multifaceted interplay of the mechanical and physicochemical influences of the durability of the adhesive seal at the composite-tooth interface. Antimicrobial peptides have been coupled into the adhesive system using non-bonded interactions [146], and subsequently, antimicrobial peptides were conjugated into the network of the adhesive system in order to improve the antimicrobials’ effectiveness [147]. An antimicrobial peptide AMP2-derivative (AMPM7) sequence using a functional spacer was used for integration into a monomer site. This adhesive system formed of co-tethered peptides demonstrated both localized calcium phosphate remineralization and strong metabolic inhibition of S. mutans [163]. An adhesive system incorporated with an antimicrobial peptide inhibited bacterial attack, and a hydroxyapatite-binding peptide promoted the remineralization of damaged tooth structures [146,163]. In 2017, Su et al. demonstrated that a cured antimicrobial peptide with nisin-incorporated dental adhesive showed a significant inhibitory effect on the growth of S. mutans [133], and recently, a paper showed that 3% (w/v) of nisin-incorporated universal adhesive system substantially inhibited the growth of both saliva-derived multispecies biofilms and S. mutans monospecific biofilms without hindering the bonding performance [166].
Moreover, it was demonstrated that C. albicans colonization on the denture’s base was significantly less than the control when histatin-adsorbed PMMA (poly methyl methacrylate), an antimicrobial peptide, was used [161]. Another report suggested that histidine-rich polypeptides were effective in the treatment of denture stomatitis [121], thus evidencing the important use of peptides in removable prostheses.
Some limitations relative to the applications of peptides in the dental field can be cited. One restriction is the absence of homogeneity of the type and obtention of the peptides used in the different applications described in the present review. Another limitation that can be highlighted is that due to the heterogeneity of the analytical techniques used for distinguishing the peptides, analyzing data using any statistical analysis was avoided.

5. Conclusions

The use of peptides has been gaining increasing attention in contemporary dentistry. Dental research evidence suggests that peptides have several applications, including osseointegration, guided tissue regeneration, vital pulp therapy, antimicrobial activity, enamel remineralization, and the surface modification of dental implants. The lack of clinical trials should be highlighted, leaving a wide space available for the investigation of peptides in dentistry.

Author Contributions

Conceptualization, L.H., R.B., Y.H. and C.E.C.-S.; methodology, L.H., R.B., N.K. and C.E.C.-S.; software, L.H., R.B. and C.E.C.-S.; validation, D.M., J.C.A.C., M.J., N.K., V.T. and Y.H.; formal analysis, L.H., R.B. and C.E.C.-S.; investigation, L.H., R.B., N.K., M.L.-S., F.S.-Y. and D.M.; resources, A.J.M.-Á., Y.H., M.J., N.K., F.S.-Y., M.L.-S. and D.M.; data curation, L.H., R.B.,Y.H. and C.E.C.-S.; writing—original draft preparation, L.H., R.B., Y.H. and C.E.C.-S.; writing—review and editing, M.L.-S., L.H., N.K. and Y.H.; visualization, N.K., A.J.M.-Á., M.J., J.C.A.C., V.T., L.H., R.B. and M.J.; supervision, L.H.; project administration, L.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on reasonable request from the first author (L.H.).

Acknowledgments

Authors L.H. and R.B. would like to acknowledge the Saint Joseph University of Beirut, Lebanon. Furthermore, the referees would also recognize the University of Hidalgo State, Mexico, the Polytechnic University of Marche, the Medical University of Lodz, and the University of Strasbourg for accompanying this research.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kharouf, N.; Haikel, Y.; Ball, V. Polyphenols in Dental Applications. Bioengineering 2020, 7, 72. [Google Scholar] [CrossRef]
  2. Hachem, C.E.; Chedid, J.C.A.; Nehme, W.; Kaloustian, M.K.; Ghosn, N.; Sahnouni, H.; Mancino, D.; Haikel, Y.; Kharouf, N. Physicochemical and Antibacterial Properties of Conventional and Two Premixed Root Canal Filling Materials in Primary Teeth. J. Funct. Biomater. 2022, 13, 177. [Google Scholar] [CrossRef] [PubMed]
  3. Kharouf, N.; Pedullà, E.; La Rosa, G.R.M.; Bukiet, F.; Sauro, S.; Haikel, Y.; Mancino, D. In Vitro Evaluation of Different Irrigation Protocols on Intracanal Smear Layer Removal in Teeth with or without Pre-Endodontic Proximal Wall Restoration. J. Clin. Med. 2020, 9, 3325. [Google Scholar] [CrossRef] [PubMed]
  4. Malta, C.P.; Barcelos, R.C.S.; Segat, H.J.; Burger, M.E.; Bier, C.A.S.; Morgental, R.D. Toxicity of Bioceramic and Resinous Endodontic Sealers Using an Alternative Animal Model: Artemia Salina. J. Conserv. Dent. 2022, 25, 185. [Google Scholar] [CrossRef]
  5. Sismanoglu, S.; Ercal, P. The Cytotoxic Effects of Various Endodontic Irrigants on the Viability of Dental Mesenchymal Stem Cells. Aust. Endod. J. 2022, 48, 305–312. [Google Scholar] [CrossRef]
  6. Bermúdez, M.; Hoz, L.; Montoya, G.; Nidome, M.; Pérez-Soria, A.; Romo, E.; Soto-Barreras, U.; Garnica-Palazuelos, J.; Aguilar-Medina, M.; Ramos-Payán, R. Bioactive Synthetic Peptides for Oral Tissues Regeneration. Front. Mater. 2021, 8, 655495. [Google Scholar] [CrossRef]
  7. Lien, S.; Lowman, H.B. Therapeutic Peptides. Trends Biotechnol. 2003, 21, 556–562. [Google Scholar] [CrossRef]
  8. Sato, A.K.; Viswanathan, M.; Kent, R.B.; Wood, C.R. Therapeutic Peptides: Technological Advances Driving Peptides into Development. Curr. Opin. Biotechnol. 2006, 17, 638–642. [Google Scholar] [CrossRef] [PubMed]
  9. Vlieghe, P.; Lisowski, V.; Martinez, J.; Khrestchatisky, M. Synthetic Therapeutic Peptides: Science and Market. Drug Discov. Today 2010, 15, 40–56. [Google Scholar] [CrossRef]
  10. Hamley, I.W. Small Bioactive Peptides for Biomaterials Design and Therapeutics. Chem. Rev. 2017, 117, 14015–14041. [Google Scholar] [CrossRef] [Green Version]
  11. Scavello, F.; Kharouf, N.; Lavalle, P.; Haikel, Y.; Schneider, F.; Metz-Boutigue, M.-H. The Antimicrobial Peptides Secreted by the Chromaffin Cells of the Adrenal Medulla Link the Neuroendocrine and Immune Systems: From Basic to Clinical Studies. Front. Immunol. 2022, in press. [CrossRef]
  12. Alkilzy, M.; Santamaria, R.; Schmoeckel, J.; Splieth, C. Treatment of Carious Lesions Using Self-Assembling Peptides. Adv. Dent. Res. 2018, 29, 42–47. [Google Scholar] [CrossRef]
  13. Fosgerau, K.; Hoffmann, T. Peptide Therapeutics: Current Status and Future Directions. Drug Discov. Today 2015, 20, 122–128. [Google Scholar] [CrossRef] [PubMed]
  14. Lau, J.L.; Dunn, M.K. Therapeutic Peptides: Historical Perspectives, Current Development Trends, and Future Directions. Bioorg. Med. Chem. 2018, 26, 2700–2707. [Google Scholar] [CrossRef]
  15. Banting, F.G.; Best, C.H.; Collip, J.B.; Campbell, W.R.; Fletcher, A.A. Pancreatic Extracts in the Treatment of Diabetes Mellitus. Can. Med. Assoc. J. 1922, 12, 141. [Google Scholar]
  16. Stürmer, E. Vasopressin, Oxytocin and Synthetic Analogues: The Use of Bioassays. J. Pharm. Biomed. Anal. 1989, 7, 199–210. [Google Scholar] [CrossRef]
  17. Dang, T.; Süssmuth, R.D. Bioactive Peptide Natural Products as Lead Structures for Medicinal Use. Acc. Chem. Res. 2017, 50, 1566–1576. [Google Scholar] [CrossRef] [PubMed]
  18. Erak, M.; Bellmann-Sickert, K.; Els-Heindl, S.; Beck-Sickinger, A.G. Peptide Chemistry Toolbox–Transforming Natural Peptides into Peptide Therapeutics. Bioorg. Med. Chem. 2018, 26, 2759–2765. [Google Scholar] [CrossRef]
  19. Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Peters, M.D.; Horsley, T.; Weeks, L. PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef] [PubMed]
  20. Bertoldi, C.; Labriola, A.; Generali, L. Dental Root Surface Treatment with Ethylenediaminetetraacetic Acid Does Not Improve Enamel Matrix Derivative Peptide Treatment within Intrabony Defects: A Retrospective Study. J. Biol. Regul. Homeost. Agents 2019, 33, 1945–1947. [Google Scholar]
  21. Hossein, B.G.; Sadr, A.; Espigares, J.; Hariri, I.; Nakashima, S.; Hamba, H.; Shafiei, F.; Moztarzadeh, F.; Tagami, J. Study on the Influence of Leucine-Rich Amelogenin Peptide (LRAP) on the Remineralization of Enamel Defects via Micro-Focus X-ray Computed Tomography and Nanoindentation. Biomed. Mater. 2015, 10, 035007. [Google Scholar] [CrossRef]
  22. Gonçalves, F.M.C.; Delbem, A.C.B.; Gomes, L.F.; Emerenciano, N.G.; Pessan, J.P.; Romero, G.D.A.; Cannon, M.L.; Danelon, M. Effect of Fluoride, Casein Phosphopeptide-Amorphous Calcium Phosphate and Sodium Trimetaphosphate Combination Treatment on the Remineralization of Caries Lesions: An in Vitro Study. Arch. Oral Biol. 2021, 122, 105001. [Google Scholar] [CrossRef]
  23. Kim, H.; Lee, W.S.; Jeong, J.; Kim, D.S.; Lee, S.; Kim, S. Effect of Elastin-like Polypeptide Incorporation on the Adhesion Maturation of Mineral Trioxide Aggregates. J. Biomed. Mater. Res. B Appl. Biomater. 2020, 108, 2847–2856. [Google Scholar] [CrossRef]
  24. Li, C.; Yang, P.; Kou, Y.; Zhang, D.; Li, M. The Polypeptide OP3-4 Induced Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells via Protein Kinase B/Glycogen Synthase Kinase 3β/β-Catenin Pathway and Promoted Mandibular Defect Bone Regeneration. Arch. Oral Biol. 2021, 130, 105243. [Google Scholar] [CrossRef]
  25. Minguela, J.; Müller, D.; Mücklich, F.; Llanes, L.; Ginebra, M.; Roa, J.; Mas-Moruno, C. Peptidic Biofunctionalization of Laser Patterned Dental Zirconia: A Biochemical-Topographical Approach. Mater. Sci. Eng. C 2021, 125, 112096. [Google Scholar] [CrossRef]
  26. Niu, J.Y.; Yin, I.X.; Wu, W.K.K.; Li, Q.-L.; Mei, M.L.; Chu, C.H. Efficacy of the Dual-Action GA-KR12 Peptide for Remineralising Initial Enamel Caries: An in Vitro Study. Clin. Oral Investig. 2022, 26, 2441–2451. [Google Scholar] [CrossRef]
  27. Pietruski, J.; Pietruska, M.; Stokowska, W.; Pattarelli, G. Evaluation of Polypeptide Growth Factors in the Process of Dental Implant Osseointegration. Rocz. Akad. Med. Bialymstoku 1995 2001, 46, 19–27. [Google Scholar]
  28. Wang, R.; Nisar, S.; Vogel, Z.; Liu, H.; Wang, Y. Dentin Collagen Denaturation Status Assessed by Collagen Hybridizing Peptide and Its Effect on Bio-Stabilization of Proanthocyanidins. Dent. Mater. 2022, 38, 748–758. [Google Scholar] [CrossRef]
  29. Yamaguchi, M.; Kojima, T.; Kanekawa, M.; Aihara, N.; Nogimura, A.; Kasai, K. Neuropeptides Stimulate Production of Interleukin-1β, Interleukin-6, and Tumor Necrosis Factor-α in Human Dental Pulp Cells. Inflamm. Res. 2004, 53, 199–204. [Google Scholar] [CrossRef]
  30. Park, C.; Song, M.; Kim, S.; Min, B. Vitronectin-Derived Peptide Promotes Reparative Dentin Formation. J. Dent. Res. 2022, 101, 1481–1489. [Google Scholar] [CrossRef]
  31. Yaguchi, A.; Hiramatsu, H.; Ishida, A.; Oshikawa, M.; Ajioka, I.; Muraoka, T. Hydrogel-Stiffening and Non-Cell Adhesive Properties of Amphiphilic Peptides with Central Alkylene Chains. Chem. Eur. J. 2021, 27, 9295–9301. [Google Scholar] [CrossRef]
  32. Xie, S.; Allington, R.W.; Svec, F.; Fréchet, J.M. Rapid Reversed-Phase Separation of Proteins and Peptides Using Optimized ‘Moulded’Monolithic Poly (Styrene–Co-Divinylbenzene) Columns. J. Chromatogr. A 1999, 865, 169–174. [Google Scholar] [CrossRef]
  33. Pastor, J.J.; Fernández, I.; Rabanal, F.; Giralt, E. A New Method for the Preparation of Unprotected Peptides on Biocompatible Resins with Application in Combinatorial Chemistry. Org. Lett. 2002, 4, 3831–3833. [Google Scholar] [CrossRef]
  34. Mizuno, M.; Goto, K.; Miura, T.; Hosaka, D.; Inazu, T. A Novel Peptide Synthesis Using Fluorous Chemistry. Chem. Commun. 2003, 8, 972–973. [Google Scholar] [CrossRef]
  35. Khurshid, Z.; Naseem, M.; Sheikh, Z.; Najeeb, S.; Shahab, S.; Zafar, M.S. Oral Antimicrobial Peptides: Types and Role in the Oral Cavity. Saudi Pharm. J. 2016, 24, 515–524. [Google Scholar] [CrossRef]
  36. Park, S.-C.; Park, Y.; Hahm, K.-S. The Role of Antimicrobial Peptides in Preventing Multidrug-Resistant Bacterial Infections and Biofilm Formation. Int. J. Mol. Sci. 2011, 12, 5971–5992. [Google Scholar] [CrossRef]
  37. Li, C.; Bhatt, P.; Johnston, T. Evaluation of a Mucoadhesive Buccal Patch for Delivery of Peptides: In Vitro Screening of Bioadhesion. Drug Dev. Ind. Pharm. 1998, 24, 919–926. [Google Scholar] [CrossRef]
  38. Kokkonen, H.; Cassinelli, C.; Verhoef, R.; Morra, M.; Schols, H.; Tuukkanen, J. Differentiation of Osteoblasts on Pectin-Coated Titanium. Biomacromolecules 2008, 9, 2369–2376. [Google Scholar] [CrossRef]
  39. Glimcher, M.; Levine, P. Studies of the Proteins, Peptides and Free Amino Acids of Mature Bovine Enamel. Biochem. J. 1966, 98, 742. [Google Scholar] [CrossRef]
  40. Takayama, S.; Kato, T.; Imamura, K.; Kita, D.; Ota, K.; Suzuki, E.; Sugito, H.; Saitoh, E.; Taniguchi, M.; Saito, A. Effect of a Mouthrinse Containing Rice Peptide CL (14-25) on Early Dental Plaque Regrowth: A Randomized Crossover Pilot Study. BMC Res. Notes 2015, 8, 531. [Google Scholar] [CrossRef]
  41. Bagno, A.; Piovan, A.; Dettin, M.; Chiarion, A.; Brun, P.; Gambaretto, R.; Fontana, G.; Di Bello, C.; Palù, G.; Castagliuolo, I. Human Osteoblast-like Cell Adhesion on Titanium Substrates Covalently Functionalized with Synthetic Peptides. Bone 2007, 40, 693–699. [Google Scholar] [CrossRef]
  42. Artzi, Z.; Weinreb, M.; Tal, H.; Nemcovsky, C.E.; Rohrer, M.D.; Prasad, H.S.; Kozlovsky, A. Experimental Intrabony and Periodontal Defects Treated with Natural Mineral Combined With a Synthetic Cell-Binding Peptide in the Canine: Morphometric Evaluations. J. Periodontol. 2006, 77, 1658–1664. [Google Scholar] [CrossRef]
  43. Bröseler, F.; Tietmann, C.; Bommer, C.; Drechsel, T.; Heinzel-Gutenbrunner, M.; Jepsen, S. Randomised Clinical Trial Investigating Self-Assembling Peptide P11-4 in the Treatment of Early Caries. Clin. Oral Investig. 2020, 24, 123–132. [Google Scholar] [CrossRef]
  44. Butz, F.; Bächle, M.; Ofer, M.; Marquardt, K.; Kohal, R.J. Sinus Augmentation with Bovine Hydroxyapatite/Synthetic Peptide in a Sodium Hyaluronate Carrier (PepGen P-15 Putty): A Clinical Investigation of Different Healing Times. Int. J. Oral Maxillofac. Implant. 2011, 26, 1317–1323. [Google Scholar]
  45. Chung, H.-Y.; Huang, K.-C. Effects of Peptide Concentration on Remineralization of Eroded Enamel. J. Mech. Behav. Biomed. Mater. 2013, 28, 213–221. [Google Scholar] [CrossRef]
  46. Altankhishig, B.; Polan, M.A.A.; Qiu, Y.; Hasan, M.R.; Saito, T. Dentin Phosphophoryn-Derived Peptide Promotes Odontoblast Differentiation In Vitro and Dentin Regeneration In Vivo. Materials 2021, 14, 874. [Google Scholar] [CrossRef] [PubMed]
  47. Afami, M.E.; El Karim, I.; About, I.; Coulter, S.M.; Laverty, G.; Lundy, F.T. Ultrashort Peptide Hydrogels Display Antimicrobial Activity and Enhance Angiogenic Growth Factor Release by Dental Pulp Stem/Stromal Cells. Materials 2021, 14, 2237. [Google Scholar] [CrossRef]
  48. Babaji, P.; Melkundi, M.; Bhagwat, P.; Mehta, V. An in Vitro Evaluation of Remineralizing Capacity of Self-Assembling Peptide (SAP) P11-4 and Casein Phosphopeptides-Amorphous Calcium Phosphate (CPP-ACP) on Artificial Enamel. Pesqui. Bras. Odontopediatr. Clín. Integr. 2019, 19, e4504. [Google Scholar] [CrossRef]
  49. Dettin, M.; Conconi, M.T.; Gambaretto, R.; Pasquato, A.; Folin, M.; Di Bello, C.; Parnigotto, P.P. Novel Osteoblast-adhesive Peptides for Dental/Orthopedic Biomaterials. J. Biomed. Mater. Res. 2002, 60, 466–471. [Google Scholar] [CrossRef] [PubMed]
  50. Cirera, A.; Manzanares, M.C.; Sevilla, P.; Ortiz-Hernandez, M.; Galindo-Moreno, P.; Gil, J. Biofunctionalization with a TGFβ-1 Inhibitor Peptide in the Osseointegration of Synthetic Bone Grafts: An in Vivo Study in Beagle Dogs. Materials 2019, 12, 3168. [Google Scholar] [CrossRef]
  51. Boda, S.K.; Almoshari, Y.; Wang, H.; Wang, X.; Reinhardt, R.A.; Duan, B.; Wang, D.; Xie, J. Mineralized Nanofiber Segments Coupled with Calcium-Binding BMP-2 Peptides for Alveolar Bone Regeneration. Acta Biomater. 2019, 85, 282–293. [Google Scholar] [CrossRef]
  52. Chen, X.; Zhou, X.; Liu, S.; Wu, R.; Aparicio, C.; Wu, J. In Vivo Osseointegration of Dental Implants with an Antimicrobial Peptide Coating. J. Mater. Sci. Mater. Med. 2017, 28, 76. [Google Scholar] [CrossRef] [PubMed]
  53. Aref, N.S.; Alrasheed, M.K. Casein Phosphopeptide Amorphous Calcium Phosphate and Universal Adhesive Resin as a Complementary Approach for Management of White Spot Lesions: An In-Vitro Study. Prog. Orthod. 2022, 23, 10. [Google Scholar] [CrossRef]
  54. Aruna, G. Estimation of N-Terminal Telopeptides of Type I Collagen in Periodontal Health, Disease and after Nonsurgical Periodontal Therapy in Gingival Crevicular Fluid: A Clinico-Biochemical Study. Indian J. Dent. Res. 2015, 26, 152. [Google Scholar] [CrossRef]
  55. Brunton, P.; Davies, R.; Burke, J.; Smith, A.; Aggeli, A.; Brookes, S.; Kirkham, J. Treatment of Early Caries Lesions Using Biomimetic Self-Assembling Peptides—A Clinical Safety Trial. Br. Dent. J. 2013, 215, E6. [Google Scholar] [CrossRef]
  56. Fang, D.; Yuran, S.; Reches, M.; Catunda, R.; Levin, L.; Febbraio, M. A Peptide Coating Preventing the Attachment of Porphyromonas Gingivalis on the Surfaces of Dental Implants. J. Periodontal Res. 2020, 55, 503–510. [Google Scholar] [CrossRef]
  57. Goldberg, A.; Advincula, M.; Komabayashi, T.; Patel, P.; Mather, P.; Goberman, D.; Kazemi, R. Polypeptide-Catalyzed Biosilicification of Dentin Surfaces. J. Dent. Res. 2009, 88, 377–381. [Google Scholar] [CrossRef] [PubMed]
  58. Amin, H.D.; Olsen, I.; Knowles, J.C.; Donos, N. Differential Effect of Amelogenin Peptides on Osteogenic Differentiation in Vitro: Identification of Possible New Drugs for Bone Repair and Regeneration. Tissue Eng. Part A 2012, 18, 1193–1202. [Google Scholar] [CrossRef] [PubMed]
  59. Godoy-Gallardo, M.; Mas-Moruno, C.; Yu, K.; Manero, J.M.; Gil, F.J.; Kizhakkedathu, J.N.; Rodriguez, D. Antibacterial Properties of HLf1–11 Peptide onto Titanium Surfaces: A Comparison Study between Silanization and Surface Initiated Polymerization. Biomacromolecules 2015, 16, 483–496. [Google Scholar] [CrossRef] [PubMed]
  60. Dommisch, H.; Skora, P.; Hirschfeld, J.; Olk, G.; Hildebrandt, L.; Jepsen, S. The Guardians of the Periodontium—Sequential and Differential Expression of Antimicrobial Peptides during Gingival Inflammation. Results from In Vivo and In Vitro Studies. J. Clin. Periodontol. 2019, 46, 276–285. [Google Scholar] [CrossRef]
  61. Dommisch, H.; Staufenbiel, I.; Schulze, K.; Stiesch, M.; Winkel, A.; Fimmers, R.; Dommisch, J.; Jepsen, S.; Miosge, N.; Adam, K. Expression of Antimicrobial Peptides and Interleukin-8 during Early Stages of Inflammation: An Experimental Gingivitis Study. J. Periodontal Res. 2015, 50, 836–845. [Google Scholar] [CrossRef]
  62. Fernandez-Garcia, E.; Chen, X.; Gutierrez-Gonzalez, C.F.; Fernandez, A.; Lopez-Esteban, S.; Aparicio, C. Peptide-Functionalized Zirconia and New Zirconia/Titanium Biocermets for Dental Applications. J. Dent. 2015, 43, 1162–1174. [Google Scholar] [CrossRef] [PubMed]
  63. Fiorellini, J.P.; Glindmann, S.; Salcedo, J.; Weber, H.-P.; Park, C.-J.; Sarmiento, H.L. The Effect of Osteopontin and an Osteopontin-Derived Synthetic Peptide Coating on Osseointegration of Implants in a Canine Model. Int. J. Periodontics Restor. Dent. 2016, 36, e88–e94. [Google Scholar] [CrossRef]
  64. Goeke, J.E.; Kist, S.; Schubert, S.; Hickel, R.; Huth, K.C.; Kollmuss, M. Sensitivity of Caries Pathogens to Antimicrobial Peptides Related to Caries Risk. Clin. Oral Investig. 2018, 22, 2519–2525. [Google Scholar] [CrossRef] [PubMed]
  65. Galler, K.M.; Hartgerink, J.D.; Cavender, A.C.; Schmalz, G.; D’Souza, R.N. A Customized Self-Assembling Peptide Hydrogel for Dental Pulp Tissue Engineering. Tissue Eng. Part A 2012, 18, 176–184. [Google Scholar] [CrossRef]
  66. Kirkham, J.; Firth, A.; Vernals, D.; Boden, N.; Robinson, C.; Shore, R.; Brookes, S.; Aggeli, A. Self-Assembling Peptide Scaffolds Promote Enamel Remineralization. J. Dent. Res. 2007, 86, 426–430. [Google Scholar] [CrossRef]
  67. Kämmerer, P.; Heller, M.; Brieger, J.; Klein, M.; Al-Nawas, B.; Gabriel, M. Immobilisation of Linear and Cyclic RGD-Peptides on Titanium Surfaces and Their Impact on Endothelial Cell Adhesion and Proliferation. Eur. Cell Mater. 2011, 21, 364–372. [Google Scholar] [CrossRef]
  68. Golland, L.; Schmidlin, P.R.; Schätzle, M. The Potential of Self-Assembling Peptides for Enhancement of in Vitro Remineralisation of White Spot Lesions as Measured by Quantitative Laser Fluorescence. Oral Health Prev. Dent. 2017, 15, 147–152. [Google Scholar] [PubMed]
  69. Hsu, C.; Chung, H.; Yang, J.-M.; Shi, W.; Wu, B. Influence of 8DSS Peptide on Nano-Mechanical Behavior of Human Enamel. J. Dent. Res. 2011, 90, 88–92. [Google Scholar] [CrossRef]
  70. Kwak, S.; Litman, A.; Margolis, H.; Yamakoshi, Y.; Simmer, J. Biomimetic Enamel Regeneration Mediated by Leucine-Rich Amelogenin Peptide. J. Dent. Res. 2017, 96, 524–530. [Google Scholar] [CrossRef]
  71. Kong, E.F.; Tsui, C.; Boyce, H.; Ibrahim, A.; Hoag, S.W.; Karlsson, A.J.; Meiller, T.F.; Jabra-Rizk, M.A. Development and in Vivo Evaluation of a Novel Histatin-5 Bioadhesive Hydrogel Formulation against Oral Candidiasis. Antimicrob. Agents Chemother. 2016, 60, 881–889. [Google Scholar] [CrossRef] [PubMed]
  72. Koch, F.; Ekat, K.; Kilian, D.; Hettich, T.; Germershaus, O.; Lang, H.; Peters, K.; Kreikemeyer, B. A Versatile Biocompatible Antibiotic Delivery System Based on Self-Assembling Peptides with Antimicrobial and Regenerative Potential. Adv. Healthc. Mater. 2019, 8, 1900167. [Google Scholar] [CrossRef] [PubMed]
  73. Hashimoto, K.; Yoshinari, M.; Matsuzaka, K.; Shiba, K.; Inoue, T. Identification of Peptide Motif That Binds to the Surface of Zirconia. Dent. Mater. J. 2011, 30, 935–940. [Google Scholar] [CrossRef] [PubMed]
  74. Kind, L.; Stevanovic, S.; Wuttig, S.; Wimberger, S.; Hofer, J.; Müller, B.; Pieles, U. Biomimetic Remineralization of Carious Lesions by Self-Assembling Peptide. J. Dent. Res. 2017, 96, 790–797. [Google Scholar] [CrossRef]
  75. Gonçalves, F.M.C.; Delbem, A.C.B.; Gomes, L.F.; Emerenciano, N.G.; dos Passos Silva, M.; Cannon, M.L.; Danelon, M. Combined Effect of Casein Phosphopeptide-Amorphous Calcium Phosphate and Sodium Trimetaphosphate on the Prevention of Enamel Demineralization and Dental Caries: An in Vitro Study. Clin. Oral Investig. 2021, 25, 2811–2820. [Google Scholar] [CrossRef]
  76. Kim, S.; Choi, J.-Y.; Jung, S.Y.; Kang, H.K.; Min, B.-M.; Yeo, I.-S.L. A Laminin-Derived Functional Peptide, PPFEGCIWN, Promotes Bone Formation on Sandblasted, Large-Grit, Acid-Etched Titanium Implant Surfaces. Int. J. Oral Maxillofac. Implant. 2019, 34, 836–844. [Google Scholar] [CrossRef] [PubMed]
  77. Kohgo, T.; Yamada, Y.; Ito, K.; Yajima, A.; Yoshimi, R.; Okabe, K.; Baba, S.; Ueda, M. Bone Regeneration with Self-Assembling Peptide Nanofiber Scaffolds in Tissue Engineering for Osseointegration of Dental Implants. Int. J. Periodontics Restor. Dent. 2011, 31, e9–e16. [Google Scholar]
  78. Gungormus, M.; Oren, E.E.; Horst, J.A.; Fong, H.; Hnilova, M.; Somerman, M.J.; Snead, M.L.; Samudrala, R.; Tamerler, C.; Sarikaya, M. Cementomimetics—Constructing a Cementum-like Biomineralized Microlayer via Amelogenin-Derived Peptides. Int. J. Oral Sci. 2012, 4, 69–77. [Google Scholar] [CrossRef] [PubMed]
  79. Kakegawa, A.; Oida, S.; Gomi, K.; Nagano, T.; Yamakoshi, Y.; Fukui, T.; Kanazashi, M.; Arai, T.; Fukae, M. Cytodifferentiation Activity of Synthetic Human Enamel Sheath Protein Peptides. J. Periodontal Res. 2010, 45, 643–649. [Google Scholar] [CrossRef]
  80. Kramer, P.R.; JanikKeith, A.; Cai, Z.; Ma, S.; Watanabe, I. Integrin Mediated Attachment of Periodontal Ligament to Titanium Surfaces. Dent. Mater. 2009, 25, 877–883. [Google Scholar] [CrossRef]
  81. Hua, J.; Yamarthy, R.; Felsenstein, S.; Scott, R.W.; Markowitz, K.; Diamond, G. Activity of Antimicrobial Peptide Mimetics in the Oral Cavity: I. Activity against Biofilms of Candida Albicans. Mol. Oral Microbiol. 2010, 25, 418–425. [Google Scholar] [CrossRef]
  82. Kohlgraf, K.G.; Ackermann, A.; Lu, X.; Burnell, K.; Bélanger, M.; Cavanaugh, J.E.; Xie, H.; Progulske-Fox, A.; Brogden, K.A. Defensins Attenuate Cytokine Responses yet Enhance Antibody Responses to Porphyromonas Gingivalis Adhesins in Mice. Future Microbiol. 2010, 5, 115–125. [Google Scholar] [CrossRef]
  83. Holmberg, K.V.; Abdolhosseini, M.; Li, Y.; Chen, X.; Gorr, S.-U.; Aparicio, C. Bio-Inspired Stable Antimicrobial Peptide Coatings for Dental Applications. Acta Biomater. 2013, 9, 8224–8231. [Google Scholar] [CrossRef] [PubMed]
  84. Koidou, V.P.; Argyris, P.P.; Skoe, E.P.; Siqueira, J.M.; Chen, X.; Zhang, L.; Hinrichs, J.E.; Costalonga, M.; Aparicio, C. Peptide Coatings Enhance Keratinocyte Attachment towards Improving the Peri-Implant Mucosal Seal. Biomater. Sci. 2018, 6, 1936–1945. [Google Scholar] [CrossRef] [PubMed]
  85. Knaup, T.; Korbmacher-Steiner, H.; Jablonski-Momeni, A. Effect of the Caries-Protective Self-Assembling Peptide P11-4 on Shear Bond Strength of Metal Brackets. J. Orofac. Orthop. Kieferorthopädie 2021, 82, 329–336. [Google Scholar] [CrossRef]
  86. Kihara, H.; Kim, D.M.; Nagai, M.; Nojiri, T.; Nagai, S.; Chen, C.-Y.; Lee, C.; Hatakeyama, W.; Kondo, H.; Da Silva, J. Epithelial Cell Adhesion Efficacy of a Novel Peptide Identified by Panning on a Smooth Titanium Surface. Int. J. Oral Sci. 2018, 10, 21. [Google Scholar] [CrossRef]
  87. Jablonski-Momeni, A.; Nothelfer, R.; Morawietz, M.; Kiesow, A.; Korbmacher-Steiner, H. Impact of Self-Assembling Peptides in Remineralisation of Artificial Early Enamel Lesions Adjacent to Orthodontic Brackets. Sci. Rep. 2020, 10, 15132. [Google Scholar] [CrossRef] [PubMed]
  88. Kamal, D.; Hassanein, H.; Elkassas, D.; Hamza, H. Comparative Evaluation of Remineralizing Efficacy of Biomimetic Self-Assembling Peptide on Artificially Induced Enamel Lesions: An in Vitro Study. J. Conserv. Dent. 2018, 21, 536. [Google Scholar] [CrossRef]
  89. Mao, B.; Xie, Y.; Yang, H.; Yu, C.; Ma, P.; You, Z.; Tsauo, C.; Chen, Y.; Cheng, L.; Han, Q. Casein Phosphopeptide-Amorphous Calcium Phosphate Modified Glass Ionomer Cement Attenuates Demineralization and Modulates Biofilm Composition in Dental Caries. Dent. Mater. J. 2021, 40, 84–93. [Google Scholar] [CrossRef]
  90. Makihira, S.; Nikawa, H.; Shuto, T.; Nishimura, M.; Mine, Y.; Tsuji, K.; Okamoto, K.; Sakai, Y.; Sakai, M.; Imari, N. 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, 2765–2772. [Google Scholar] [CrossRef]
  91. Li, Q.-L.; Ning, T.-Y.; Cao, Y.; Zhang, W.; Mei, M.L.; Chu, C.H. A Novel Self-Assembled Oligopeptide Amphiphile for Biomimetic Mineralization of Enamel. BMC Biotechnol. 2014, 14, 32. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, Z.; Ma, S.; Duan, S.; Xuliang, D.; Sun, Y.; Zhang, X.; Xu, X.; Guan, B.; Wang, C.; Hu, M. 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, 5124–5136. [Google Scholar] [CrossRef]
  93. Min, S.-K.; Kang, H.K.; Jang, D.H.; Jung, S.Y.; Kim, O.B.; Min, B.-M.; Yeo, I.-S. Titanium Surface Coating with a Laminin-Derived Functional Peptide Promotes Bone Cell Adhesion. BioMed Res. Int. 2013, 2013, 638348. [Google Scholar] [CrossRef]
  94. Moore, A.; Perez, S.; Hartgerink, J.; D’Souza, R.; Colombo, J. Ex Vivo Modeling of Multidomain Peptide Hydrogels with Intact Dental Pulp. J. Dent. Res. 2015, 94, 1773–1781. [Google Scholar] [CrossRef] [PubMed]
  95. Muruve, N.; Feng, Y.; Platnich, J.; Hassett, D.; Irvin, R.; Muruve, D.; Cheng, F. A Peptide-Based Biological Coating for Enhanced Corrosion Resistance of Titanium Alloy Biomaterials in Chloride-Containing Fluids. J. Biomater. Appl. 2017, 31, 1225–1234. [Google Scholar] [CrossRef]
  96. Nguyen, P.K.; Gao, W.; Patel, S.D.; Siddiqui, Z.; Weiner, S.; Shimizu, E.; Sarkar, B.; Kumar, V.A. Self-Assembly of a Dentinogenic Peptide Hydrogel. ACS Omega 2018, 3, 5980–5987. [Google Scholar] [CrossRef] [PubMed]
  97. Mardas, N.; Stavropoulos, A.; Karring, T. Calvarial Bone Regeneration by a Combination of Natural Anorganic Bovine-derived Hydroxyapatite Matrix Coupled with a Synthetic Cell-binding Peptide (PepGenTM): An Experimental Study in Rats. Clin. Oral Implant. Res. 2008, 19, 1010–1015. [Google Scholar] [CrossRef]
  98. Mateescu, M.; Baixe, S.; Garnier, T.; Jierry, L.; Ball, V.; Haikel, Y.; Metz-Boutigue, M.H.; Nardin, M.; Schaaf, P.; Etienne, O. Antibacterial Peptide-Based Gel for Prevention of Medical Implanted-Device Infection. PLoS ONE 2015, 10, e0145143. [Google Scholar] [CrossRef] [PubMed]
  99. Liu, Y.; Fan, L.; Lin, X.; Zou, L.; Li, Y.; Ge, X.; Fu, W.; Zhang, Z.; Xiao, K.; Lv, H. Functionalized Self-Assembled Peptide RAD/Dentonin Hydrogel Scaffold Promotes Dental Pulp Regeneration. Biomed. Mater. 2021, 17, 015009. [Google Scholar] [CrossRef] [PubMed]
  100. Li, Y.; Wang, Y.; Chen, X.; Jiang, W.; Jiang, X.; Zeng, Y.; Li, X.; Feng, Z.; Luo, J.; Zhang, L. Antimicrobial Peptide GH12 as Root Canal Irrigant Inhibits Biofilm and Virulence of Enterococcus Faecalis. Int. Endod. J. 2020, 53, 948–961. [Google Scholar] [CrossRef]
  101. Mancino, D.; Kharouf, N.; Scavello, F.; Hellé, S.; Salloum-Yared, F.; Mutschler, A.; Mathieu, E.; Lavalle, P.; Metz-Boutigue, M.-H.; Haïkel, Y. The Catestatin-Derived Peptides Are New Actors to Fight the Development of Oral Candidosis. Int. J. Mol. Sci. 2022, 23, 2066. [Google Scholar] [CrossRef] [PubMed]
  102. Mai, S.; Mauger, M.T.; Niu, L.; Barnes, J.B.; Kao, S.; Bergeron, B.E.; Ling, J.; Tay, F.R. Potential Applications of Antimicrobial Peptides and Their Mimics in Combating Caries and Pulpal Infections. Acta Biomater. 2017, 49, 16–35. [Google Scholar] [CrossRef]
  103. Lv, X.; Yang, Y.; Han, S.; Li, D.; Tu, H.; Li, W.; Zhou, X.; Zhang, L. Potential of an Amelogenin Based Peptide in Promoting Reminerlization of Initial Enamel Caries. Arch. Oral Biol. 2015, 60, 1482–1487. [Google Scholar] [CrossRef]
  104. Lee, J.-Y.; Choo, J.-E.; Choi, Y.-S.; Park, J.-B.; Min, D.-S.; Lee, S.-J.; Rhyu, H.K.; Jo, I.-H.; Chung, C.-P.; Park, Y.-J. Assembly of Collagen-Binding Peptide with Collagen as a Bioactive Scaffold for Osteogenesis in Vitro and in Vivo. Biomaterials 2007, 28, 4257–4267. [Google Scholar] [CrossRef]
  105. Liang, K.; Xiao, S.; Liu, H.; Shi, W.; Li, J.; Gao, Y.; He, L.; Zhou, X.; Li, J. 8DSS Peptide Induced Effective Dentinal Tubule Occlusion in Vitro. Dent. Mater. 2018, 34, 629–640. [Google Scholar] [CrossRef]
  106. Lee, S.J.; Won, J.-E.; Han, C.; Yin, X.Y.; Kim, H.K.; Nah, H.; Kwon, I.K.; Min, B.-H.; Kim, C.-H.; Shin, Y.S. Development of a Three-Dimensionally Printed Scaffold Grafted with Bone Forming Peptide-1 for Enhanced Bone Regeneration with in Vitro and in Vivo Evaluations. J. Colloid Interface Sci. 2019, 539, 468–480. [Google Scholar] [CrossRef] [PubMed]
  107. Na, D.H.; Faraj, J.; Capan, Y.; Leung, K.P.; DeLuca, P.P. Chewing Gum of Antimicrobial Decapeptide (KSL) as a Sustained Antiplaque Agent: Preformulation Study. J. Control. Release 2005, 107, 122–130. [Google Scholar] [CrossRef] [PubMed]
  108. Magalhães, G.d.A.P.; Fraga, M.A.A.; de Souza Araújo, I.J.; Pacheco, R.R.; Correr, A.B.; Puppin-Rontani, R.M. Effect of a Self-Assembly Peptide on Surface Roughness and Hardness of Bleached Enamel. J. Funct. Biomater. 2022, 13, 79. [Google Scholar] [CrossRef] [PubMed]
  109. Lallier, T.E.; Palaiologou, A.A.; Yukna, R.A.; Layman, D.L. The Putative Collagen-binding Peptide P-15 Promotes Fibroblast Attachment to Root Shavings but Not Hydroxyapatite. J. Periodontol. 2003, 74, 458–467. [Google Scholar] [CrossRef] [PubMed]
  110. Li, M.; Yang, Y.; Lin, C.; Zhang, Q.; Gong, L.; Wang, Y.; Zhang, X. Antibacterial Properties of Small-Size Peptide Derived from Penetratin against Oral Streptococci. Materials 2021, 14, 2730. [Google Scholar] [CrossRef]
  111. Matsugishi, A.; Aoki-Nonaka, Y.; Yokoji-Takeuchi, M.; Yamada-Hara, M.; Mikami, Y.; Hayatsu, M.; Terao, Y.; Domon, H.; Taniguchi, M.; Takahashi, N. Rice Peptide with Amino Acid Substitution Inhibits Biofilm Formation by Porphyromonas Gingivalis and Fusobacterium Nucleatum. Arch. Oral Biol. 2021, 121, 104956. [Google Scholar] [CrossRef]
  112. Li, X.; Yu, Z.; Jiang, S.; Dai, X.; Wang, G.; Wang, Y.; Yang, Z.; Gao, J.; Zou, H. An Amelogenin-Based Peptide Hydrogel Promoted the Odontogenic Differentiation of Human Dental Pulp Cells. Regen. Biomater. 2022, 9, rbac039. [Google Scholar]
  113. Mishra, P.R.N.; Kolte, A.P.; Kolte, R.A.; Pajnigara, N.G.; Shah, K.K. Comparative Evaluation of Open Flap Debridement Alone and in Combination with Anorganic Bone Matrix/Cell-Binding Peptide in the Treatment of Human Infrabony Defects: A Randomized Clinical Trial. J. Indian Soc. Periodontol. 2019, 23, 42. [Google Scholar]
  114. Padovano, J.; Ravindran, S.; Snee, P.; Ramachandran, A.; Bedran-Russo, A.; George, A. DMP1-Derived Peptides Promote Remineralization of Human Dentin. J. Dent. Res. 2015, 94, 608–614. [Google Scholar] [PubMed]
  115. Park, J.H.; Gillispie, G.J.; Copus, J.S.; Zhang, W.; Atala, A.; Yoo, J.J.; Yelick, P.C.; Lee, S.J. The Effect of BMP-Mimetic Peptide Tethering Bioinks on the Differentiation of Dental Pulp Stem Cells (DPSCs) in 3D Bioprinted Dental Constructs. Biofabrication 2020, 12, 035029. [Google Scholar]
  116. Pellissari, C.V.G.; Jorge, J.H.; Marin, L.M.; Sabino-Silva, R.; Siqueira, W.L. Statherin-Derived Peptides as Antifungal Strategy against Candida Albicans. Arch. Oral Biol. 2021, 125, 105106. [Google Scholar] [CrossRef]
  117. Petzold, C.; Monjo, M.; Rubert, M.; Reinholt, F.P.; Gomez-Florit, M.; Ramis, J.M.; Ellingsen, J.E.; Lyngstadaas, S.P. Effect of Proline-Rich Synthetic Peptide--Coated Titanium Implants on Bone Healing in a Rabbit Model. Oral Craniofac. Tissue Eng. 2012, 2, 35–43. [Google Scholar] [CrossRef]
  118. Picker, A.; Nicoleau, L.; Nonat, A.; Labbez, C.; Cölfen, H. Identification of Binding Peptides on Calcium Silicate Hydrate: A Novel View on Cement Additives. Adv. Mater. 2014, 26, 1135–1140. [Google Scholar] [CrossRef] [PubMed]
  119. Pihl, M.; Galli, S.; Jimbo, R.; Andersson, M. Osseointegration and Antibacterial Effect of an Antimicrobial Peptide Releasing Mesoporous Titania Implant. J. Biomed. Mater. Res. B Appl. Biomater. 2021, 109, 1787–1795. [Google Scholar] [PubMed]
  120. Ren, Q.; Li, Z.; Ding, L.; Wang, X.; Niu, Y.; Qin, X.; Zhou, X.; Zhang, L. Anti-Biofilm and Remineralization Effects of Chitosan Hydrogel Containing Amelogenin-Derived Peptide on Initial Caries Lesions. Regen. Biomater. 2018, 5, 69–76. [Google Scholar]
  121. Santarpia, R.P., III; Pollock, J.J.; Renner, R.P.; Gwinnett, A.J. In Vivo Antifungal Efficacy of Salivary Histidine-Rich Polypeptides: Preliminary Findings in a Denture Stomatitis Model System. J. Prosthet. Dent. 1991, 66, 693–699. [Google Scholar] [CrossRef] [PubMed]
  122. Schmidlin, P.; Zobrist, K.; Attin, T.; Wegehaupt, F. In Vitro Re-Hardening of Artificial Enamel Caries Lesions Using Enamel Matrix Proteins or Self-Assembling Peptides. J. Appl. Oral Sci. 2016, 24, 31–36. [Google Scholar] [CrossRef]
  123. Schmitt, C.M.; Koepple, M.; Moest, T.; Neumann, K.; Weisel, T.; Schlegel, K.A. In Vivo Evaluation of Biofunctionalized Implant Surfaces with a Synthetic Peptide (P-15) and Its Impact on Osseointegration. A Preclinical Animal Study. Clin. Oral Implant. Res. 2016, 27, 1339–1348. [Google Scholar] [CrossRef] [PubMed]
  124. Schuler, M.; Owen, G.R.; Hamilton, D.W.; de Wild, M.; Textor, M.; Brunette, D.M.; Tosatti, S.G. Biomimetic Modification of Titanium Dental Implant Model Surfaces Using the RGDSP-Peptide Sequence: A Cell Morphology Study. Biomaterials 2006, 27, 4003–4015. [Google Scholar] [CrossRef] [PubMed]
  125. Schuster, L.; Ardjomandi, N.; Munz, M.; Umrath, F.; Klein, C.; Rupp, F.; Reinert, S.; Alexander, D. Establishment of Collagen: Hydroxyapatite/BMP-2 Mimetic Peptide Composites. Materials 2020, 13, 1203. [Google Scholar] [CrossRef] [PubMed]
  126. Secchi, A.G.; Grigoriou, V.; Shapiro, I.M.; Cavalcanti-Adam, E.A.; Composto, R.J.; Ducheyne, P.; Adams, C.S. RGDS Peptides Immobilized on Titanium Alloy Stimulate Bone Cell Attachment, Differentiation and Confer Resistance to Apoptosis. J. Biomed. Mater. Res. Part Off. J. Soc. Biomater. Jpn. Soc. Biomater. Aust. Soc. Biomater. Korean Soc. Biomater. 2007, 83, 577–584. [Google Scholar] [CrossRef] [PubMed]
  127. Segvich, S.J.; Smith, H.C.; Kohn, D.H. The Adsorption of Preferential Binding Peptides to Apatite-Based Materials. Biomaterials 2009, 30, 1287–1298. [Google Scholar] [CrossRef]
  128. Sfeir, C.; Fang, P.-A.; Jayaraman, T.; Raman, A.; Xiaoyuan, Z.; Beniash, E. Synthesis of Bone-like Nanocomposites Using Multiphosphorylated Peptides. Acta Biomater. 2014, 10, 2241–2249. [Google Scholar] [CrossRef]
  129. Shi, J.; Liu, Y.; Wang, Y.; Zhang, J.; Zhao, S.; Yang, G. Biological and Immunotoxicity Evaluation of Antimicrobial Peptide-Loaded Coatings Using a Layer-by-Layer Process on Titanium. Sci. Rep. 2015, 5, 16336. [Google Scholar] [CrossRef] [Green Version]
  130. Shinkai, K.; Taira, Y.; Suzuki, M.; Kato, C.; Yamauchi, J.; Suzuki, S.; Katoh, Y. Dentin Bond Strength of an Experimental Adhesive System Containing Calcium Chloride, Synthetic Peptides Derived from Dentin Matrix Protein 1 (PA and PB), and Hydroxyapatite for Direct Pulp Capping and as a Bonding Agent. Odontology 2010, 98, 110–116. [Google Scholar] [CrossRef]
  131. Shinkai, K.; Taira, Y.; Suzuki, M.; Kato, C.; Yamauchi, J.; Suzuki, S.; Katoh, Y. Effect of the Concentrations of Calcium Chloride and Synthetic Peptides in Primers on Dentin Bond Strength of an Experimental Adhesive System. Dent. Mater. J. 2010, 29, 738–746. [Google Scholar] [CrossRef] [PubMed]
  132. Shuturminska, K.; Tarakina, N.V.; Azevedo, H.S.; Bushby, A.J.; Mata, A.; Anderson, P.; Al-Jawad, M. Elastin-like Protein, with Statherin Derived Peptide, Controls Fluorapatite Formation and Morphology. Front. Physiol. 2017, 8, 368. [Google Scholar] [CrossRef] [PubMed]
  133. Su, M.; Yao, S.; Gu, L.; Huang, Z.; Mai, S. Antibacterial Effect and Bond Strength of a Modified Dental Adhesive Containing the Peptide Nisin. Peptides 2018, 99, 189–194. [Google Scholar] [CrossRef]
  134. Suaid, F.A.; Macedo, G.O.; Novaes, A.B., Jr.; Borges, G.J.; Souza, S.L.; Taba, M., Jr.; Palioto, D.B.; Grisi, M.F. The Bone Formation Capabilities of the Anorganic Bone Matrix–Synthetic Cell-Binding Peptide 15 Grafts in an Animal Periodontal Model: A Histologic and Histomorphometric Study in Dogs. J. Periodontol. 2010, 81, 594–603. [Google Scholar] [CrossRef] [PubMed]
  135. Sugawara, S.; Maeno, M.; Lee, C.; Nagai, S.; Kim, D.M.; Da Silva, J.; Nagai, M.; Kondo, H. Establishment of Epithelial Attachment on Titanium Surface Coated with Platelet Activating Peptide. PLoS ONE 2016, 11, e0164693. [Google Scholar] [CrossRef] [PubMed]
  136. Sun, X.; Huang, X.; Tan, X.; Si, Y.; Wang, X.; Chen, F.; Zheng, S. Salivary Peptidome Profiling for Diagnosis of Severe Early Childhood Caries. J. Transl. Med. 2016, 14, 240. [Google Scholar] [CrossRef]
  137. Takahashi, N.; Sato, T. Dipeptide Utilization by the Periodontal Pathogens Porphyromonas Gingivalis, Prevotella Intermedia, Prevotella Nigrescens and Fusobacterium Nucleatum. Oral Microbiol. Immunol. 2002, 17, 50–54. [Google Scholar] [CrossRef]
  138. Tanhaieian, A.; Pourgonabadi, S.; Akbari, M.; Mohammadipour, H.-S. The Effective and Safe Method for Preventing and Treating Bacteria-Induced Dental Diseases by Herbal Plants and a Recombinant Peptide. J. Clin. Exp. Dent. 2020, 12, e523. [Google Scholar] [CrossRef]
  139. Üstün, N.; Aktören, O. Analysis of Efficacy of the Self-assembling Peptide-based Remineralization Agent on Artificial Enamel Lesions. Microsc. Res. Tech. 2019, 82, 1065–1072. [Google Scholar] [CrossRef]
  140. Wang, B.; Wu, B.; Jia, Y.; Jiang, Y.; Yuan, Y.; Man, Y.; Xiang, L. Neural Peptide Promotes the Angiogenesis and Osteogenesis around Oral Implants. Cell. Signal. 2021, 79, 109873. [Google Scholar] [CrossRef]
  141. Wang, D.; Shen, Y.; Hancock, R.E.; Ma, J.; Haapasalo, M. Antimicrobial Effect of Peptide DJK-5 Used Alone or Mixed with EDTA on Mono-and Multispecies Biofilms in Dentin Canals. J. Endod. 2018, 44, 1709–1713. [Google Scholar] [CrossRef]
  142. Warnke, P.H.; Voss, E.; Russo, P.A.; Stephens, S.; Kleine, M.; Terheyden, H.; Liu, Q. Antimicrobial Peptide Coating of Dental Implants: Biocompatibility Assessment of Recombinant Human Beta Defensin-2 for Human Cells. Int. J. Oral Maxillofac. Implant. 2013, 28, 982–988. [Google Scholar] [CrossRef] [PubMed]
  143. Werner, S.; Huck, O.; Frisch, B.; Vautier, D.; Elkaim, R.; Voegel, J.-C.; Brunel, G.; Tenenbaum, H. The Effect of Microstructured Surfaces and Laminin-Derived Peptide Coatings on Soft Tissue Interactions with Titanium Dental Implants. Biomaterials 2009, 30, 2291–2301. [Google Scholar] [CrossRef]
  144. Winfred, S.B.; Meiyazagan, G.; Panda, J.J.; Nagendrababu, V.; Deivanayagam, K.; Chauhan, V.S.; Venkatraman, G. Antimicrobial Activity of Cationic Peptides in Endodontic Procedures. Eur. J. Dent. 2014, 8, 254–260. [Google Scholar] [CrossRef]
  145. Wu, J.; Mao, S.; Xu, L.; Qiu, D.; Wang, S.; Dong, Y. Odontogenic Differentiation Induced by TGF-Β1 Binding Peptide–Modified Bioglass. J. Dent. Res. 2022, 101, 1190–1197. [Google Scholar] [CrossRef]
  146. Xie, S.-X.; Boone, K.; VanOosten, S.K.; Yuca, E.; Song, L.; Ge, X.; Ye, Q.; Spencer, P.; Tamerler, C. Peptide Mediated Antimicrobial Dental Adhesive System. Appl. Sci. 2019, 9, 557. [Google Scholar] [CrossRef]
  147. Xie, S.-X.; Song, L.; Yuca, E.; Boone, K.; Sarikaya, R.; VanOosten, S.K.; Misra, A.; Ye, Q.; Spencer, P.; Tamerler, C. Antimicrobial Peptide–Polymer Conjugates for Dentistry. ACS Appl. Polym. Mater. 2020, 2, 1134–1144. [Google Scholar] [CrossRef]
  148. Yamamoto, N.; Maeda, H.; Tomokiyo, A.; Fujii, S.; Wada, N.; Monnouchi, S.; Kono, K.; Koori, K.; Teramatsu, Y.; Akamine, A. Expression and Effects of Glial Cell Line-derived Neurotrophic Factor on Periodontal Ligament Cells. J. Clin. Periodontol. 2012, 39, 556–564. [Google Scholar] [CrossRef]
  149. Yamashita, M.; Lazarov, M.; Jones, A.A.; Mealey, B.L.; Mellonig, J.T.; Cochran, D.L. Periodontal Regeneration Using an Anabolic Peptide with Two Carriers in Baboons. J. Periodontol. 2010, 81, 727–736. [Google Scholar] [CrossRef] [PubMed]
  150. Yang, X.; Huang, P.; Wang, H.; Cai, S.; Liao, Y.; Mo, Z.; Xu, X.; Ding, C.; Zhao, C.; Li, J. Antibacterial and Anti-Biofouling Coating on Hydroxyapatite Surface Based on Peptide-Modified Tannic Acid. Colloids Surf. B Biointerfaces 2017, 160, 136–143. [Google Scholar] [CrossRef] [PubMed]
  151. Yang, X.; Li, Z.; Xiao, H.; Wang, N.; Li, Y.; Xu, X.; Chen, Z.; Tan, H.; Li, J. A Universal and Ultrastable Mineralization Coating Bioinspired from Biofilms. Adv. Funct. Mater. 2018, 28, 1802730. [Google Scholar] [CrossRef]
  152. Yang, X.; Yang, B.; He, L.; Li, R.; Liao, Y.; Zhang, S.; Yang, Y.; Xu, X.; Zhang, D.; Tan, H. Bioinspired Peptide-Decorated Tannic Acid for in Situ Remineralization of Tooth Enamel: In Vitro and in Vivo Evaluation. ACS Biomater. Sci. Eng. 2017, 3, 3553–3562. [Google Scholar] [CrossRef] [PubMed]
  153. Yang, X.; Zhang, D.; Liu, G.; Wang, J.; Luo, Z.; Peng, X.; Zeng, X.; Wang, X.; Tan, H.; Li, J. Bioinspired from Mussel and Salivary Acquired Pellicle: A Universal Dual-Functional Polypeptide Coating for Implant Materials. Mater. Today Chem. 2019, 14, 100205. [Google Scholar] [CrossRef]
  154. Yang, Y.; Xia, L.; Haapasalo, M.; Wei, W.; Zhang, D.; Ma, J.; Shen, Y. A Novel Hydroxyapatite-Binding Antimicrobial Peptide against Oral Biofilms. Clin. Oral Investig. 2019, 23, 2705–2712. [Google Scholar] [CrossRef] [PubMed]
  155. Yang, Y.; Yang, B.; Li, M.; Wang, Y.; Yang, X.; Li, J. Salivary Acquired Pellicle-Inspired DpSpSEEKC Peptide for the Restoration of Demineralized Tooth Enamel. Biomed. Mater. 2017, 12, 025007. [Google Scholar] [CrossRef]
  156. Yang, Z.; Liu, M.; Yang, Y.; Zheng, M.; Liu, X.; Tan, J. Biofunctionalization of Zirconia with Cell-Adhesion Peptides via Polydopamine Crosslinking for Soft Tissue Engineering: Effects on the Biological Behaviors of Human Gingival Fibroblasts and Oral Bacteria. RSC Adv. 2020, 10, 6200–6212. [Google Scholar] [CrossRef]
  157. Yazici, H.; Fong, H.; Wilson, B.; Oren, E.; Amos, F.; Zhang, H.; Evans, J.; Snead, M.; Sarikaya, M.; Tamerler, C. Biological Response on a Titanium Implant-Grade Surface Functionalized with Modular Peptides. Acta Biomater. 2013, 9, 5341–5352. [Google Scholar] [CrossRef]
  158. Ye, Q.; Spencer, P.; Yuca, E.; Tamerler, C. Engineered Peptide Repairs Defective Adhesive–Dentin Interface. Macromol. Mater. Eng. 2017, 302, 1600487. [Google Scholar] [CrossRef]
  159. Ye, W.; Yeghiasarian, L.; Cutler, C.W.; Bergeron, B.E.; Sidow, S.; Xu, H.H.; Niu, L.; Ma, J.; Tay, F.R. Comparison of the Use of D-Enantiomeric and l-Enantiomeric Antimicrobial Peptides Incorporated in a Calcium-Chelating Irrigant against Enterococcus Faecalis Root Canal Wall Biofilms. J. Dent. 2019, 91, 103231. [Google Scholar] [CrossRef]
  160. Yonehara, N.; Shibutani, T.; Tsai, H.-Y.; Inoki, R. Effects of Opioids and Opioid Peptide on the Release of Substance P-like Material Induced by Tooth Pulp Stimulation in the Trigeminal Nucleus Caudalis of the Rabbit. Eur. J. Pharmacol. 1986, 129, 209–216. [Google Scholar] [CrossRef]
  161. Yoshinari, M.; Kato, T.; Matsuzaka, K.; Hayakawa, T.; Inoue, T.; Oda, Y.; Okuda, K.; Shimono, M. Adsorption Behavior of Antimicrobial Peptide Histatin 5 on PMMA. J. Biomed. Mater. Res. Part B Appl. Biomater. Off. J. Soc. Biomater. Jpn. Soc. Biomater. Aust. Soc. Biomater. Korean Soc. Biomater. 2006, 77, 47–54. [Google Scholar] [CrossRef]
  162. Yoshinari, M.; Kato, T.; Matsuzaka, K.; Hayakawa, T.; Shiba, K. Prevention of Biofilm Formation on Titanium Surfaces Modified with Conjugated Molecules Comprised of Antimicrobial and Titanium-Binding Peptides. Biofouling 2010, 26, 103–110. [Google Scholar] [CrossRef]
  163. Yuca, E.; Xie, S.-X.; Song, L.; Boone, K.; Kamathewatta, N.; Woolfolk, S.K.; Elrod, P.; Spencer, P.; Tamerler, C. Reconfigurable Dual Peptide Tethered Polymer System Offers a Synergistic Solution for Next Generation Dental Adhesives. Int. J. Mol. Sci. 2021, 22, 6552. [Google Scholar] [CrossRef] [PubMed]
  164. Zhang, P.; Wu, S.; Li, J.; Bu, X.; Dong, X.; Chen, N.; Li, F.; Zhu, J.; Sang, L.; Zeng, Y. Dual-Sensitive Antibacterial Peptide Nanoparticles Prevent Dental Caries. Theranostics 2022, 12, 4818. [Google Scholar] [CrossRef]
  165. Zhang, T.; Wang, Z.; Hancock, R.E.; De La Fuente-Núñez, C.; Haapasalo, M. Treatment of Oral Biofilms by a D-Enantiomeric Peptide. PLoS ONE 2016, 11, e0166997. [Google Scholar] [CrossRef]
  166. Zhao, M.; Qu, Y.; Liu, J.; Mai, S.; Gu, L. A Universal Adhesive Incorporating Antimicrobial Peptide Nisin: Effects on Streptococcus Mutans and Saliva-Derived Multispecies Biofilms. Odontology 2020, 108, 376–385. [Google Scholar] [CrossRef] [PubMed]
  167. Zhou, B.; Liu, Y.; Wei, W.; Mao, J. GEPIs-HA Hybrid: A Novel Biomaterial for Tooth Repair. Med. Hypotheses 2008, 71, 591–593. [Google Scholar] [CrossRef] [PubMed]
  168. Zhou, L.; Lai, Y.; Huang, W.; Huang, S.; Xu, Z.; Chen, J.; Wu, D. Biofunctionalization of Microgroove Titanium Surfaces with an Antimicrobial Peptide to Enhance Their Bactericidal Activity and Cytocompatibility. Colloids Surf. B Biointerfaces 2015, 128, 552–560. [Google Scholar] [CrossRef]
  169. Gungormus, M.; Tulumbaci, F. Peptide-Assisted Pre-Bonding Remineralization of Dentin to Improve Bonding. J. Mech. Behav. Biomed. Mater. 2021, 113, 104119. [Google Scholar] [CrossRef]
  170. Gug, H.R.; Park, Y.-H.; Park, S.-J.; Jang, J.Y.; Lee, J.-H.; Lee, D.-S.; Shon, W.-J.; Park, J.-C. Novel Strategy for Dental Caries by Physiologic Dentin Regeneration with CPNE7 Peptide. Arch. Oral Biol. 2022, 143, 105531. [Google Scholar] [CrossRef] [PubMed]
  171. Murray, C.J.; Vos, T.; Lozano, R.; Naghavi, M.; Flaxman, A.D.; Michaud, C.; Ezzati, M.; Shibuya, K.; Salomon, J.A.; Abdalla, S. Disability-Adjusted Life Years (DALYs) for 291 Diseases and Injuries in 21 Regions, 1990–2010: A Systematic Analysis for the Global Burden of Disease Study 2010. Lancet 2012, 380, 2197–2223. [Google Scholar] [CrossRef]
  172. González-Cabezas, C. The Chemistry of Caries: Remineralization and Demineralization Events with Direct Clinical Relevance. Dent. Clin. 2010, 54, 469–478. [Google Scholar] [CrossRef] [PubMed]
  173. Innes, N.; Schwendicke, F. Restorative Thresholds for Carious Lesions: Systematic Review and Meta-Analysis. J. Dent. Res. 2017, 96, 501–508. [Google Scholar] [CrossRef] [PubMed]
  174. Dirks, O.B. Posteruptive Changes in Dental Enamel. J. Dent. Res. 1966, 45, 503–511. [Google Scholar] [CrossRef]
  175. Da Pontte, A.C.A.; Damião, A.O.M.C.; Rosa, A.M.; Da Silva, A.N.; Fachin, A.V.; Cortecazzi, A., Jr.; Marinho, A.L.D.; Prudente, A.C.L.; Pulgas, A.T.; Machado, A.D.; et al. Consensus Guidelines for the Management of Inflammatory Bowel Disease. Arq. Gastroenterol. 2010, 47, 313–325. [Google Scholar] [CrossRef]
  176. Gao, S.S.; Zhang, S.; Mei, M.L.; Lo, E.C.-M.; Chu, C.-H. Caries Remineralisation and Arresting Effect in Children by Professionally Applied Fluoride Treatment–a Systematic Review. BMC Oral Health 2016, 16, 12. [Google Scholar] [CrossRef]
  177. Lenzi, T.L.; Montagner, A.F.; Soares, F.Z.M.; de Oliveira Rocha, R. Are Topical Fluorides Effective for Treating Incipient Carious Lesions? A Systematic Review and Meta-Analysis. J. Am. Dent. Assoc. 2016, 147, 84–91. [Google Scholar] [CrossRef]
  178. Lussi, A.; Hellwig, E.; Klimek, J. Fluorides—Mode of Action and Recommendations for Use. Schweiz. Monatsschr. Zahnmed. 2012, 122, 1030. [Google Scholar]
  179. Alkilzy, M.; Tarabaih, A.; Santamaria, R.; Splieth, C. Self-Assembling Peptide P11-4 and Fluoride for Regenerating Enamel. J. Dent. Res. 2018, 97, 148–154. [Google Scholar] [CrossRef] [PubMed]
  180. Aggeli, A.; Bell, M.; Boden, N.; Carrick, L.M.; Strong, A.E. Self-Assembling Peptide Polyelectrolyte Β-Sheet Complexes Form Nematic Hydrogels. Angew. Chem. 2003, 115, 5761–5764. [Google Scholar] [CrossRef]
  181. Aggeli, A.; Bell, M.; Boden, N.; Keen, J.; Knowles, P.; McLeish, T.; Pitkeathly, M.; Radford, S. Responsive Gels Formed by the Spontaneous Self-Assembly of Peptides into Polymeric β-Sheet Tapes. Nature 1997, 386, 259–262. [Google Scholar] [CrossRef] [PubMed]
  182. Davies, R.; Aggeli, A.; Beevers, A.; Boden, N.; Carrick, L.; Fishwick, C.; McLeish, T.; Nyrkova, I.; Semenov, A. Self-Assembling β-Sheet Tape Forming Peptides. Supramol. Chem. 2006, 18, 435–443. [Google Scholar] [CrossRef]
  183. Kyle, S.; Aggeli, A.; Ingham, E.; McPherson, M.J. Recombinant Self-Assembling Peptides as Biomaterials for Tissue Engineering. Biomaterials 2010, 31, 9395–9405. [Google Scholar] [CrossRef]
  184. Dabdoub, S.; Tsigarida, A.; Kumar, P. Patient-Specific Analysis of Periodontal and Peri-Implant Microbiomes. J. Dent. Res. 2013, 92, 168S–175S. [Google Scholar] [CrossRef] [PubMed]
  185. Arciniegas, M.; Aparicio, C.; Manero, J.; Gil, F. Low Elastic Modulus Metals for Joint Prosthesis: Tantalum and Nickel–Titanium Foams. J. Eur. Ceram. Soc. 2007, 27, 3391–3398. [Google Scholar] [CrossRef]
  186. Branemark, P.-I. Osseointegrated Implants in the Treatment of the Edentulous Jaw. Experience from a 10-Year Period. Scand. J. Plast. Reconstr. Surg. Suppl. 1977, 16, 1–132. [Google Scholar]
  187. Schwartz-Arad, D.; Kidron, N.; Dolev, E. A Long-term Study of Implants Supporting Overdentures as a Model for Implant Success. J. Periodontol. 2005, 76, 1431–1435. [Google Scholar] [CrossRef]
  188. Lindquist, L.; Carlsson, G.; Jemt, T. A Prospective 15-year Follow-up Study of Mandibular Fixed Prostheses Supported by Osseointegrated Implants. Clinical Results and Marginal Bone Loss. Clin. Oral Implant. Res. 1996, 7, 329–336. [Google Scholar] [CrossRef]
  189. Bumgardner, J.D.; Adatrow, P.; Haggard, W.O.; Norowski, P.A. Emerging Antibacterial Biomaterial Strategies for the Prevention of Peri-Implant Inflammatory Diseases. Int. J. Oral Maxillofac. Implant. 2011, 26, 553–560. [Google Scholar]
  190. Le Guéhennec, L.; Soueidan, A.; Layrolle, P.; Amouriq, Y. Surface Treatments of Titanium Dental Implants for Rapid Osseointegration. Dent. Mater. 2007, 23, 844–854. [Google Scholar] [CrossRef]
  191. Schliephake, H.; Scharnweber, D. Chemical and Biological Functionalization of Titanium for Dental Implants. J. Mater. Chem. 2008, 18, 2404–2414. [Google Scholar] [CrossRef]
  192. Chen, X.; Li, Y.; Aparicio, C.; Nazarpour, S.; Chaker, M. Thin Films and Coatings in Biology; Biological and Medical Physics–Biomedical Engineering Series; Springer: Berlin/Heidelberg, Germany, 2013. [Google Scholar]
  193. Neu, H.C. The Crisis in Antibiotic Resistance. Science 1992, 257, 1064–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Abdolhosseini, M.; Nandula, S.R.; Song, J.; Hirt, H.; Gorr, S.-U. Lysine Substitutions Convert a Bacterial-Agglutinating Peptide into a Bactericidal Peptide That Retains Anti-Lipopolysaccharide Activity and Low Hemolytic Activity. Peptides 2012, 35, 231–238. [Google Scholar] [CrossRef]
  195. Hirt, H.; Gorr, S.-U. Antimicrobial Peptide GL13K Is Effective in Reducing Biofilms of Pseudomonas Aeruginosa. Antimicrob. Agents Chemother. 2013, 57, 4903–4910. [Google Scholar] [CrossRef]
  196. Bhatnagar, R.S.; Qian, J.J.; Wedrychowska, A.; Sadeghi, M.; Wu, Y.M.; Smith, N. Design of Biomimetic Habitats for Tissue Engineering with P-15, a Synthetic Peptide Analogue of Collagen. Tissue Eng. 1999, 5, 53–65. [Google Scholar] [CrossRef] [PubMed]
  197. Steindorff, M.M.; Lehl, H.; Winkel, A.; Stiesch, M. Innovative Approaches to Regenerate Teeth by Tissue Engineering. Arch. Oral Biol. 2014, 59, 158–166. [Google Scholar] [CrossRef] [PubMed]
  198. Wang, H.; Wang, S.; Cheng, L.; Jiang, Y.; Melo, M.A.S.; Weir, M.D.; Oates, T.W.; Zhou, X.; Xu, H.H. Novel Dental Composite with Capability to Suppress Cariogenic Species and Promote Non-Cariogenic Species in Oral Biofilms. Mater. Sci. Eng. C 2019, 94, 587–596. [Google Scholar] [CrossRef] [PubMed]
  199. Pinheiro, S.L.; Azenha, G.R.; Araujo, G.; Puppin Rontani, R. Effectiveness of Casein Phosphopeptide-Amorphous Calcium Phosphate and Lysozyme, Lactoferrin, and Lactoperoxidase in Reducing Streptococcus Mutans. Gen. Dent. 2017, 65, 47–50. [Google Scholar]
  200. Divyapriya, G.; Yavagal, P.C.; Veeresh, D. Casein Phosphopeptide-Amorphous Calcium Phosphate in Dentistry: An Update. Int. J. Oral Health Sci. 2016, 6, 18. [Google Scholar] [CrossRef]
  201. Hardan, L.; Bourgi, R.; Kharouf, N.; Mancino, D.; Zarow, M.; Jakubowicz, N.; Haikel, Y.; Cuevas-Suárez, C.E. Bond Strength of Universal Adhesives to Dentin: A Systematic Review and Meta-Analysis. Polymers 2021, 13, 814. [Google Scholar] [CrossRef]
  202. Elizalde-Hernández, A.; Hardan, L.; Bourgi, R.; Isolan, C.P.; Moreira, A.G.; Zamarripa-Calderón, J.E.; Piva, E.; Cuevas-Suárez, C.E.; Devoto, W.; Saad, A.; et al. Effect of Different Desensitizers on Shear Bond Strength of Self-Adhesive Resin Cements to Dentin. Bioengineering 2022, 9, 372. [Google Scholar] [CrossRef]
  203. Rodríguez, F.; Glawe, D.D.; Naik, R.R.; Hallinan, K.P.; Stone, M.O. Study of the Chemical and Physical Influences upon in Vitro Peptide-Mediated Silica Formation. Biomacromolecules 2004, 5, 261–265. [Google Scholar] [CrossRef] [PubMed]
  204. Branco, M.C.; Schneider, J.P. Self-Assembling Materials for Therapeutic Delivery. Acta Biomater. 2009, 5, 817–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Gao, J.; Tang, C.; Elsawy, M.A.; Smith, A.M.; Miller, A.F.; Saiani, A. Controlling Self-Assembling Peptide Hydrogel Properties through Network Topology. Biomacromolecules 2017, 18, 826–834. [Google Scholar] [CrossRef] [PubMed]
  206. Spencer, P.; Ye, Q.; Song, L.; Parthasarathy, R.; Boone, K.; Misra, A.; Tamerler, C. Threats to Adhesive/Dentin Interfacial Integrity and next Generation Bio-enabled Multifunctional Adhesives. J. Biomed. Mater. Res. B Appl. Biomater. 2019, 107, 2673–2683. [Google Scholar] [CrossRef]
  207. Hardan, L.; Lukomska-Szymanska, M.; Zarow, M.; Cuevas-Suárez, C.E.; Bourgi, R.; Jakubowicz, N.; Sokolowski, K.; D’Arcangelo, C. One-Year Clinical Aging of Low Stress Bulk-Fill Flowable Composite in Class II Restorations: A Case Report and Literature Review. Coatings. 2021, 11, 504. [Google Scholar] [CrossRef]
  208. Hardan, L.; Bourgi, R.; Cuevas-Suárez, C.E.; Zarow, M.; Kharouf, N.; Mancino, D.; Villares, C.F.; Skaba, D.; Lukomska-Szymanska, M. The Bond Strength and Anti-bacterial Activity of the Universal Dentin Bonding System: A Systematic Review and Meta- Analysis. Microorganisms 2021, 9, 1230. [Google Scholar] [CrossRef]
  209. Bourgi, R.; Daood, U.; Bijle, M.N.; Fawzy, A.; Ghaleb, M.; Hardan, L. Reinforced Universal Adhesive by Ribose Crosslinker: A Novel Strategy in Adhesive Dentistry. Polymers 2021, 13, 704. [Google Scholar] [CrossRef]
Figure 1. Flowchart summarizing the selection process of articles.
Figure 1. Flowchart summarizing the selection process of articles.
Bioengineering 10 00214 g001
Table 1. Search strategy used in the MEDLINE database.
Table 1. Search strategy used in the MEDLINE database.
(Peptide) OR (Polypeptides) OR (Polypeptide) AND (Materials, Dental) OR (Dental Material) OR (Material, Dental)
Table 2. Characteristics of the included studies.
Table 2. Characteristics of the included studies.
Study and YearType of StudyPeptide UsedApplicationMain Results
Bagno, 2007 [41]In vitroTwo adhesive peptides:
an RGD-containing peptide and a peptide recorded on human vitronectin
Implant osseointegrationIt was observed that there was a capacity of the peptides to promote enhanced cell adhesion
Artzi, 2006 [42]Experimental studyA synthetic peptide (P-15)Guided tissue regeneration and guided bone regeneration techniquesThe use of a synthetic peptide showed increased osteoconductive and biocompatible features
Bröseler, 2020 [43]Randomized clinical trialSelf-assembling peptide (SAP) P11-4Early buccal carious lesionsSelf-assembling peptide regenerated enamel caries lesions
Butz, 2011 [44]Prospective in vivo studySynthetic Peptide in a Sodium Hyaluronate Carrier (PepGen P-15 Putty)Sinus graftingThe peptide evaluated was successful for maxillary sinus augmentation
Chung, 2013 [45]In vitroAsparagine–serine–serine (NSS) peptide.Remineralization of eroded enamel.Peptide increased the nanohardness and elastic modulus of eroded enamel
Altankhishig, 2021 [46]In vitro and in vivoPeptideVital pulp therapyThe dentin phosphophoryn-derived arginine-glycine-aspartic acid-containing peptide showed adequate properties as a bioactive material for dentin regeneration
Afami, 2021 [47]In vitroUltrashort peptide hydrogel, (naphthalene-2-ly)-acetyl-diphenylalanine-dilysine-OH (NapFFεKεK-OH)Antimicrobial activity and angiogenic growth factor release by dental pulp stem/stromal cellsPeptide-containing hydrogels have potential in tissue engineering for pulp regeneration
Babaji, 2019 [48]In vitroSAP P11-4 and casein phosphopeptides-amorphous calcium phosphate (CPP-ACP)Enamel remineralizationThe peptide was more effective and efficient when compared to CPP-ACP
Dettin, 2002 [49]In vitroNovel osteoblast-adhesive peptidesOsteoblast adhesionThe novel peptide promotes proteoglycan-mediated osteoblast adhesion efficiently
Cirera, 2019 [50]In vivoTGF-β1 inhibitor peptide: P144Osseointegration of synthetic bone graftsThe healing period of osseointegrated biomaterials can be shortened when peptide biofunctionalization is used
Boda, 2020 [51]In vitroMineralized nanofiber segments combined with calcium-binding bone morphogenetic protein 2 (BMP-2)-mimicking peptidesAlveolar bone regenerationMineralized nanofibers functionalized with peptides have the potential to regenerate craniofacial bone defects
Chen, 2017 [52]In vivoGL13K-peptideOsseointegration of implantsThis study showed that titanium dental implants with an antimicrobial GL13K peptide coating enables in vivo implant osseointegration
Aref, 2022 [53]In vitroCPP-ACPWhite spot lesionCPP-ACP could be a promising approach to manage WSLs efficiently, with subsequent universal adhesive resin infiltration
Aruna, 2015 [54]ClinicalGingival crevicular fluid (GCF) N-terminal telopeptides of type I collagen (NTx)Periodontal therapyCross-linked NTx can be successfully estimated in the GCF of chronic periodontitis subjects
Brunton, 2013 [55]A clinical trialBiomimetic SAP: P11-4Early caries lesionsTreatment of early caries lesions with P11-4 is safe, and a single application of this peptide is associated with significant enamel regeneration
Fang, 2020 [56]In vitroTwo hexapeptide coatingsDental implantsThe novel hexapeptide coating can inhibit the attachment of Porphyromonas gingivalis and prevent the formation of dental biofilm
Goldberg, 2009 [57]In vitroPolypeptideOccluding dentin tubulesPeptide catalysts that mediate mineral formation can retain functionality on dentin, suggesting a wide range of preventive and treatment strategies
Amin, 2012 [58]In vitroAmelogenin PeptidesOsteogenic differentiationAmelogenin-derived peptide could be a useful tool for limiting pathological bone cell growth
Godoy-Gallardo, 2015 [59]In vitrohLf1-11 PeptideAntibacterial properties on titanium surfacesA greater amount of peptide attached to the surfaces functionalized via atom transfer radical polymerization than those functionalized via silane
Dommisch, 2019 [60]In vivo and in vitroAntimicrobial peptidesGingival inflammationThe study delivers evidence on the role of antimicrobial peptides as guardians of a healthy periodontium
Dommisch, 2015 [61]Experimental studyAntimicrobial peptidesGingivitisDifferential temporal expression for antimicrobial peptides could guarantee continuous antimicrobial activity alongside changes in the bacterial composition of the growing dental biofilm
Fernandez-Garcia, 2015 [62]In vitroPeptide-functionalized zirconiaImplantSurface bioactivation of zirconia-containing constituents for dental implant applications will allow their perfected clinical implementation by incorporating signaling oligopeptides to accelerate osseointegration, improve mucosal sealing, and/or incorporate antimicrobial properties to avoid peri-implant infections
Fiorellini, 2016 [63]In vitroOsteopontin-derived synthetic peptide: OC-1016Osseointegration of implantsOC-1016 was capable of meaningfully accelerating the initial stage of osseointegration and bone healing around implants
Goeke, 2018 [64]ClinicalAntimicrobial peptidesCaries riskThe incidence of low-susceptible strains to antimicrobial peptides appears to relate to individual caries status
Galler, 2012 [65]In vitroSAP hydrogelDental pulp tissue engineeringThe use of this innovative biomaterial was considered a highly favorable candidate for upcoming treatment hypotheses in regenerative endodontics
Kirkham, 2007 [66]In situSAP scaffoldsEnamel remineralizationSAP might be useful for dental tissue engineering
Kämmerer, 2011 [67]In vitroRGD peptidesDental implantsModifications of titanium surfaces with c-RGD peptides are an encouraging way to endorse endothelial cell growth
Golland, 2017 [68]In vitroSAPRemineralization of white spot lesionsThe application of SAP on demineralized bovine enamel indicated an irregular crystal or a lack of remineralization
Hsu, 2010 [69]In vitroAspartate-serine-serine (8DSS) pep- tidesNucleation of calcium phosphate carbonate from free ions8DSS peptides reduced the surface roughness of demineralized enamel and promoted the uniform deposition of nano-crystalline calcium phosphate carbonate over demineralized enamel surfaces
Kwak, 2017 [70]In vitroLeucine-rich amelogenin peptide (LRAP)Enamel regenerationLRAP has the power to enhance the linear growth of mature enamel crystals
Kong, 2015 [71]In vivoHistatin-5 (Hst-5)Oral CandidiasisHst-5 was able to clear existing lesions
Koch, 2019 [72]In vitroSAP:
P11-4 and P11-28/29
Periodontal therapySAP hydrogels were effective for periodontal therapy
Hashimoto, 2011 [73]In vitroPeptide motifZirconiaA peptide motif was successful in binding zirconia
Kind, 2017 [74]In vitroSAP: P11-4Remineralization of carious lesionsThe application of P11-4 might facilitate the subsurface regeneration of the enamel lesion
Gonçalves, 2020 [75]In vitroCasein phosphopeptide-amorphous calcium phosphate (MI Paste Plus)Enamel demineralization and dental cariesMI Paste Plus might be effective in improving oral health
Kim, 2019 [76]In vitro and in vivoA laminin-derived functional peptideImplantPeptide DN3 promotes bone healing
Kohgo, 2011 [77]In vitroSAPImplantSAP could be useful for bone regeneration around dental implants
Gungormus, 2012 [78]Ex vivoAmelogenin-derived peptidesPeriodontal tissuesAmelogenin-derived peptide 5 promoted the regeneration of periodontal tissues
Kakegawa, 2010 [79]In vitroEnamel sheath protein peptidesConstruction of the enamel sheath during tooth developmentA specific peptide sequence encourages the cytodifferentiation and mineralization activity of human periodontal ligaments
Kramer, 2009 [80]In vitroIntegrin blocking peptideTitanium surfacesAntibody and peptide treatment reduced the number of fibroblast cells involved on the implant surfaces
Hua, 2010 [81]In vitroAntimicrobial peptideOral cavityThe antimicrobial peptide was demonstrated as an anti-Candida agent
Hua, 2010 [81]In vitroAntimicrobial peptideOral cavityThe antimicrobial peptide exhibits potent activity against both A. actinomycetemcomitans and P. gingivalis biofilms
Kohlgraf, 2010 [82]In vitroHuman neutrophil peptide α-defensins (HNPs)Cytokine responsesThe ability of HNPs to attenuate proinflammatory cytokines was dependent upon both the defensin and antigen of P. gingivalis
Holmberg, 2013 [83]In vitroAntimicrobial peptide: GL13KDental and orthopedic implantsThe antimicrobial activity and cytocompatibility of GL13K-biofunctionalized titanium make it a promising candidate for sustained inhibition of bacterial biofilm growth
Koidou, 2019 [84]In vitroBioinspired peptide coatingsPeri-implant mucosal SealPeptide coatings were considered a promising candidate for inducing a peri-mucosal seal around dental implants
Knaup, 2021 [85]In vitroSAP: P11-4Metal bracketsThe application of the caries-protective SAP P11-4 before the bonding of brackets did not influence the shear bond strength
Kihara, 2018 [86]In vitroNovel synthetic peptide (A10)Titanium surfaceThe novel peptide has a useful presentation that might enhance advanced clinical outcomes by means of titanium implants and abutments by preventing or reducing peri-implant disease
Jablonski-Momeni, 2020 [87]In vitroSAP P11-4Early enamel lesions
adjacent to orthodontic brackets
The application of p11-4 with fluoride varnish was demonstrated to be superior for the remineralization of enamel adjacent to brackets when compared to the use of fluorides alone
Kamal, 2018 [88]In vitroSAP P11-4Artificially induced enamel lesionsSAP confers a higher remineralizing efficacy
Mao, 2021 [89]In vitroCPP-ACPDental cariesThe use of 5% CPP-ACP reduced 39% of bacterial biofilm
Makihira, 2011 [90]In vivoAntimicrobial peptide derived from histatin: JH8194Dental implantJH8194 might deliver a viable biological modification of titanium surfaces to amplify trabecular bone formation around dental implants
Li, 2014 [91]In vitroSynthetic and self-assembled oligopeptide amphiphile (OPA)Mineralization of enamelOPA was successful in the biomimetic mineralization of demineralized enamel
Liu, 2016 [92]Experimental Chimeric peptides comprising antimicrobial and titanium-binding motifsBiofilm formationChimeric peptides provide a promising alternative to inhibit the formation of biofilms on titanium surfaces with the power to prevent peri-implantitis
Min, 2013 [93]In vitroLaminin-derived functional peptide, Ln2-P3ImplantAn Ln2-P3-coated implant surface enhances bone cell adhesion
Moore, 2015 [94]Ex vivoMultidomain peptide hydrogelsDental pulpMultidomain peptide hydrogels offered centrally and peripherally within whole dental pulp tissue are demonstrated to be biocompatible and preserve the architecture of the local tissue
Muruve, 2017 [95]In vitroPEGylated metal-binding peptide (D-K122-4-PEG)Titanium surfaceD-K122-4-PEG promotes resistance to corrosion
Nguyen, 2018 [96]In vitroDentinogenic peptideDental
pulp stem cells
The SAP promised guided dentinogenesis
Mardas, 2007 [97]An experimental study in ratsPepGenBone regenerationThe anorganic bovine-derived hydroxyapatite matrix coupled with a synthetic cell-binding peptide failed to promote new bone formation
Mateescu, 2015 [98]In vitroAntimicrobial peptide CateslytinPeri-implant diseasesThe new peptide could be ideal in the prevention of peri-implant diseases
Liu, 2021 [99]In vitroRADA16-I: (SAP)Pulp regenerationThe novel SAP could be ideal in endodontic tissue engineering
Li, 2020 [100]In vitroGH12: antimicrobial peptideRoot canal irrigantGH12 suppressed E. faecali in dentinal tubules
Mancino, 2022 [101]In vitroCatestatin-derived peptidesOral candidiasisThe catestatin-derived peptides were considered for the treatment of oral candidiasis
Mai, 2016 [102]In vitroAntimicrobial peptidesCaries and pulpal infectionsAntimicrobial peptide mimics offer opportunities for new therapeutics in regenerative endodontics and root canal treatments
Lv, 2015 [103]In vitroAmelogenin based peptideRemineralization of initial enamel cariesThe amelogenin-based peptide enhances enamel caries remineralization
Lee, 2007 [104]In vitro and in vivoCollagen-binding peptideOsteogenesisCollagen-binding peptide induced biomineralization of bone
Liang, 2018 [105]In vitro8DSS peptideDentinal tubule occlusion8DSS peptide induced strong dentinal tubule occlusion and can be used in dentin hypersensitivity
Lee, 2018 [106]In vitro and in vivoBone formation peptide-1 (BFP1)Bone regenerationBFP1 was considered promising for bone repair
Na, 2005 [107]Preformulation studyAntimicrobial decapeptide (KSL)Antiplaque agentKSL served as a novel antiplaque agent in the oral cavity
Magalhães, 2022 [108]In vitroSelf-assembly peptide: P11-4Bleached enamelThe use of P11-4 after bleaching results in the fastest recovery to baseline enamel properties
Lallier, 2003 [109]In vitroCollagen-binding peptide P-15Periodontal treatmentP-15 promoted fibroblast attachment to root surfaces
Li, 2021 [110]In vitroSmall-size peptide: RR9Oral streptococciRR9 might be considered a possible antimicrobial agent for periodontal disease
Matsugishi, 2021 [111]In vitroRice peptideBiofilm formationRice peptide hindered the biofilm formation of F. nucleatum and P. gingivalis
Li, 2022 [112]In vitroAmelogenin-based peptide hydrogelHuman dental pulp cellsThe amelogenin peptide hydrogel enhanced mineralization and encouraged odontogenic differentiation
Mishra, 2019 [113]A randomized clinical trialAnorganic bone matrix/cell-binding peptide (ABM/P-15)Human infrabony periodontal defectsThe combination of ABM/P-15 was established to be a favorable material for periodontal regeneration
Padovano, 2015 [114]In vitroDMP1-derived peptidesRemineralization of human dentinDMP1-derived peptides could be useful in modulating mineral deposition
Park, 2020 [115]In vitroBMP-mimetic peptideDental pulp stem cellsBMP-mimetic peptide accelerated human dental pulp stem cells
Pellissari, 2021 [116]In vitroStatherin-derived peptidesBiofilm developmentThe natural peptides from statherin are able to decrease biofilm proliferation and Candida albicans colonization
Petzold, 2012 [117]In vivoProline-rich synthetic peptideTitanium implantsProline-rich peptides have a probable biocompatible capacity for endorsing osseointegration by lessening bone resorption
Picker, 2014 [118]In vitroBinding peptidesCalcium silicate hydrateA new strong calcium silicate hydrate-binding additive influenced the physical properties of cement
Pihl, 2021 [119]In vivoAntimicrobial peptide: RRP9W4NTitania implantRRP9W4N was demonstrated to be successful in the control of infection in osseointegrating implants
Ren, 2018 [120]In vitroChitosan hydrogel containing amelogenin-derived peptideInitial caries lesionsChitosan hydrogel containing amelogenin-derived peptide was demonstrated to be effective in controlling caries and promoting the remineralization of the initial enamel carious lesion
Santarpia, 1991 [121]In vivoHistidine-rich polypeptidesDenture stomatitisHistidine-rich polypeptides were effective in the treatment of denture stomatitis
Schmidlin, 2015 [122]In vitroSAPMineralization of artificial caries lesionsSAP improved the hardness profile of deep demineralized artificial lesions
Schmitt, 2016 [123]In vivoSynthetic peptide (P-15)OsseointegrationThere is no advantage in the early phase of osseointegration for dental implants with P-15-containing surfaces
Schuler, 2006 [124]In vitroRGDSP-peptide sequenceTitanium dental implantThere is no communication between RGD-peptide surface density and surface topography for osteoblasts
Schuster, 2020 [125]In vitroHydroxyapatite/BMP-2 mimetic peptideBone tissue engineeringBiofunctionalization of collagen-hydroxyapatite composites with BMP-2 simulated peptides was considered cost-effective and fast for prolonged and improved jaw periosteal cell proliferation
Secchi, 2007 [126]In vitroArginine-glycine-aspartic acid (RGDS) peptidesImplantThe modification of the titanium surface with RGDS peptides promoted osseointegration
Segvich, 2009 [127]In vitroBinding peptide sequencesBone regenerationThe binding peptide sequences can be used in dentin and bone
tissue engineering
Sfeir, 2014 [128]In vitroMultiphosphorylated peptidesMineralized collagen fibrils of bone and dentinUsing phosphopeptides, there is progress in biomimetic nanostructured materials for mineralized tissue regeneration and repair
Shi, 2015 [129]In vitroAntimicrobial peptide-loaded coatingsDental implantThe antimicrobial peptide-loaded coatings were demonstrated to be a potential approach for preventing peri-implantitis
Shinkai, 2010 [130]In vitroSynthetic peptides derived from dentin matrix protein 1 (pA and pB)Direct pulp capping and bonding agentThe primer containing synthetic peptides derived from dentin matrix protein 1 negatively affected the bond strength to dentin
Shinkai, 2010 [131]In vitroSynthetic peptides (pA and pB)Bonding agentA significant difference was seen in bond strength among CaCl2 concentrations in Primer-I (comprising 10 wt.% CaCl2) and pA/pB concentrations in Primer-II comprising 10 wt.% pA/pB, and there is a noteworthy interaction between these two factors
Shuturminska, 2017 [132]In vitroStatherin-derived peptideEnamel biomineralizationThe use of statherin-derived peptide was considered effective in enamel therapy
Su, 2017 [133]In vitroPeptide nisinDental adhesiveThe cured nisin included in the dental adhesive showed a noteworthy inhibitory effect on the growth of S. mutans
Suaid, 2010 [134]Histologic and histomorphometric studyAnorganic bone matrix–synthetic cell-binding peptide 15Periodontal class III furcation defectsThe use of anorganic bone matrix–synthetic cell-binding peptide 15 was effective in bone formation
Sugawara, 2016 [135]In vitroPlatelet-activating peptideTitanium surfaceAn epithelial basement membrane was formed on the titanium surface when platelet activating peptide was used
Sun, 2016 [136]ClinicalPeptidomeEarly childhood cariesThe magnetic bead-founded matrix-assisted laser desorption/ionization time-of-flight mass spectrometry was considered an effective technique for screening distinctive peptides from the saliva of junior patients with early childhood caries
Takahashi, 2002 [137]In vitroDipeptide: aspartylaspartate and glutamylglutamatePeriodontal pathogensDipeptides can be employed as growth substrates for P. intermedia, P. gingivalis, F. nucleatum, and P. nigrescens
Tanhaieian, 2020 [138]In vitroRecombinant peptideDental diseasesThe recombinant peptide was demonstrated effective as an antimicrobial agent against E. faecalis and oral streptococci
Üstuün, 2019 [139]In vitroSAP: P11-4Artificial enamel lesionsP11-4 was demonstrated to have the best remineralization efficacy
Wag, 2020 [140]In vivoNeural peptideAngiogenesis and osteogenesis around oral implantsAlpha-calcitonin gene-related peptide up-regulated the expression of Hippo-YAP and downstream genes in order to encourage osteogenesis and angiogenesis around the implants
Wang, 2015 [141]In vitroPeptide DJK-5Dentin canalsThe peptide DJK-5 showed an imperative antibacterial property against mono- and multispecies biofilms in dentin canals
Warnke, 2013 [142]In vitroHuman beta-defensins (HBDs), small cationic antimicrobial peptidesDental implantsHBD-2 is not only biocompatible with but further encourages the proliferation of human mesenchymal stem cells
Wener, 2009 [143]In vitroLaminin-derived peptideDental implantsLaminin-derived peptide improved and enhanced the integration of soft tissue on titanium implants used in dentistry
Winfred, 2014 [144]In vitroCationic peptidesEndodontic proceduresCationic peptides prevented the spread of endodontic infections
Wu, 2022 [145]In vitroTGF-β1 binding peptide–modified bioglassEndodontic therapyTGF-β1 binding peptide–modified bioglass was effective for regeneration in endodontic therapy
Xue Xie, 2019 [146]In vitroAntimicrobial peptideDental adhesive systemAntimicrobial peptide-hydrophilic adhesive delivers an advanced adhesive/dentin interface
Xue Xie, 2020 [147]In vitroAntimicrobial peptideDental adhesive systemPeptide-conjugated dentin adhesives were effective in secondary caries treatment and improved the durability of dental composites
Yakufu, 2020 [147]In vitroOsteogenic growth peptide (OGP)Osteogenesis activityOGP was promising in dental and orthopedic applications
Yamamoto, 2012 [148]In vivoPeptide including Arg-Gly-Asp (RGD) sequencePeriodontal ligament cellsGlial cell line-derived neurotrophic factor, which was hindered by pre-treatment with the peptide-embracing Arg-Gly-Asp (RGD) sequence, enhanced the appearance of bone sialoprotein and fibronectin on human periodontal ligament cells
Yamashita, 2010 [149]In vitroAnabolic peptidePeriodontal regenerationAnabolic peptide has a positive influence on bone cells
Yang, 2017 [150]In vitroPeptide-modified tannic acidHydroxyapatite surfacePeptide-modified tannic acid inhibited the adhesion of bacteria
Yang, 2018 [151]In vitroSalivary acquired pellicle (SAPe)-inspired peptide DDDEEKBiofilmsSAPe-inspired peptide DDDEEK has a great advantage in the field of implant materials
Yang, 2017 [152]In vitro and in vivoBioinspired peptide-decorated tannic acidRemineralization of tooth enamelBioinspired peptide-decorated tannic acid has a good influence on the remineralization of tooth enamel
Yang, 2019 [153]In vitroDual-functional polypeptide
Implant materialsDual-functional polypeptide has a potential application in the treatment of hard tissue-related diseases
Yang, 2019 (b) [154]In vitro and in vivoImmunomodulatory peptide 1018Plaque biofilmsImmunomodulatory peptide 1018 was effective as an anti-biofilm agent
Yang, 2017 (b) [155]In vitroDpSpSEEKC peptideDemineralized tooth enamel DpSpSEEKC restored demineralized tooth enamel
Yang, 2020 [156]In vitroCell-adhesion peptides via polydopamine crosslinkingZirconia abutment surfacesCell-adhesion peptides improved soft tissue integration around zirconia abutments via polydopamine crosslinking
Yazici, 2013 [157]In vitroModular peptides Titanium implantModular peptides on titanium surfaces improved the bioactivity of fibroblast and osteoblast cells on implant-grade materials
Ye, 2017 [158]In vitroPeptide-based approachAdhesive-dentin interfaceThe peptide-based remineralization approach was effective in designing integrated tissue-biomaterial interfaces
Ye, 2019 [159]In vitroD-enantiomeric and L-enantiomeric antimicrobial peptidesRoot canal wall biofilms D-enantiomeric peptides exhibited more antimicrobial potent activity than L-enantiomeric peptides against E. faecalis biofilms on the canal space
Yonehara, 1986 [160]In vivoOpioids and opioid peptidesTooth pulp stimulationThere is an interaction between substance P and enkephalin systems in the superficial layer of the brain-stem trigeminal sensory nuclear complex for the regulation of dental pain transmission. In addition, the native application of naloxone (5 × 10−7 M) partly antagonized the inhibitory effects of locally applied morphine and the opioid peptide
Yoshinari, 2005 [161]In vitroAntimicrobial peptide histatin 5Poly (methyl methacrylate) denture baseC. albicans colonization on histatin-adsorbed PMMA was knowingly less than the control
Yoshinari, 2010 [162]
In vitroAntimicrobial and titanium-binding
peptides
Titanium
surfaces
Antimicrobial and titanium-binding peptides were encouraging for the reduction of biofilm formation on titanium surfaces
Yuca, 2021 [163]In vitroDual-peptide tethered polymer systemDental adhesivesThe adhesive system formed of co-tethered peptides revealed both localized calcium phosphate remineralization and strong metabolic inhibition of S. mutans
Zhang, 2022 [164]In-vitroDual-sensitive antibacterial peptideDental cariesThis peptide prevented damage from bacteria and, thus, from dental caries
Zhang, 2016 [165]In vitroD-Enantiomeric peptideOral biofilmsD-enantiomeric peptide was effective against oral biofilms
Zhao, 2020 [166]In vitroAntimicrobial peptide nisinDental adhesive3% (w/v) nisin-incorporated Single Bond Universal substantially inhibited the development of both saliva-derived multispecies biofilms and monospecific S. mutans biofilms without hindering the bonding performance
Zhou, 2008 [167]In vitroGenetically engineered peptides for inorganicsTooth repairGenetically engineered peptides for inorganics were effective in tooth repair
Zhou, 2015 [168]In vitroAntimicrobial peptideTitanium surfacesAntimicrobial peptide provided a promising bifunctional surface
Gungormus, 2021 [169]In vitroPeptide-assisted pre-bondingRemineralization of dentinPre-bonding remineralization of dentin using peptide during 10 min notably enhanced the stiffness of dentin and the resistance to hydrolysis. In addition, it can reduce shrinkage due to drying
Koidou, 2018 [84]In vitroLaminin 332- and ameloblastin-derived peptides (Lam, Ambn)Peri-implant mucosal sealLaminin 332- and ameloblastin-derived peptides were demonstrated to be effective in producing a peri-mucosal seal around dental implants
Gug, 2022 [170]In vivoCPNE7-derived functional peptideDentin regeneration of dental cariesCPNE7-derived functional peptide repaired caries by dentin regeneration
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hardan, L.; Chedid, J.C.A.; Bourgi, R.; Cuevas-Suárez, C.E.; Lukomska-Szymanska, M.; Tosco, V.; Monjarás-Ávila, A.J.; Jabra, M.; Salloum-Yared, F.; Kharouf, N.; et al. Peptides in Dentistry: A Scoping Review. Bioengineering 2023, 10, 214. https://doi.org/10.3390/bioengineering10020214

AMA Style

Hardan L, Chedid JCA, Bourgi R, Cuevas-Suárez CE, Lukomska-Szymanska M, Tosco V, Monjarás-Ávila AJ, Jabra M, Salloum-Yared F, Kharouf N, et al. Peptides in Dentistry: A Scoping Review. Bioengineering. 2023; 10(2):214. https://doi.org/10.3390/bioengineering10020214

Chicago/Turabian Style

Hardan, Louis, Jean Claude Abou Chedid, Rim Bourgi, Carlos Enrique Cuevas-Suárez, Monika Lukomska-Szymanska, Vincenzo Tosco, Ana Josefina Monjarás-Ávila, Massa Jabra, Fouad Salloum-Yared, Naji Kharouf, and et al. 2023. "Peptides in Dentistry: A Scoping Review" Bioengineering 10, no. 2: 214. https://doi.org/10.3390/bioengineering10020214

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop