REVIEW ARTICLE

Therapeutic effect of marine bioactive substances against periodontitis based on in vitro, in vivo, and clinical studies

Tae-Hee Kim1,2https://orcid.org/0000-0001-7818-1946, Se-Chang Kim2,3https://orcid.org/0000-0002-6399-9726, Won-Kyo Jung1,2,3,*https://orcid.org/0000-0003-3438-8843
Author Information & Copyright
1Research Center for Marine-Integrated Bionics Technology, Pukyong National University, Busan 48513, Korea
2Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Korea
3Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-Senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Korea
*Corresponding author: Won-Kyo Jung, Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-Senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Korea, Tel: +82-51-629-5775, Fax: +82-51-629-5775, E-mail:wkjung@pknu.ac.kr

Copyright © 2022 The Korean Society of Fisheries and Aquatic Science. This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Aug 19, 2022; Revised: Sep 28, 2022; Accepted: Nov 05, 2022

Published Online: Jan 31, 2023

Abstract

Marine bioactive substances (MBS), such as phlorotannins, collagens, peptides, sterols, and polysaccharides, are increasing attention as therapeutic agents for several diseases due to their pharmacological effects. Previous studies have demonstrated the biological activities of MBS including antibacterial, anticoagulant, antidiabetic, antimicrobial, anti-inflammatory activities. Among numerous human diseases, periodontitis is one of the high-prevalence inflammatory diseases in the world. To treat periodontitis, several surgeries (bone grafting, flap surgery, and soft tissue graft) are usually used. However, the surgery for patients with chronic periodontitis induces several side effects, including additional inflammatory responses at the operated site, chronic wound healing, and secondary surgery. Therefore, this review assessed the most recent trends in MBS using Google Scholar, PubMed, and Web of Science search engines to develop marine-derived therapeutic agents for periodontitis. Further, we summarized the current applications and therapeutic potential of MBS to serve as a reference for developing novel technologies applied to MBS against periodontitis treatment.

Keywords: Marine organisms; Bioactive substance; Periodontitis

Introduction

Periodontitis is a common chronic multifactorial inflammatory disease in the periodontal tissues and is one of the high-prevalence chronic conditions in the world (Eke et al., 2020; Papapanou & Susin, 2017; Papapanou et al., 2018). Periodontitis usually occurs at supporting structures of the teeth such as alveolar bone, gingiva, periodontal ligament, and root cementum and is associated with dental plaque biofilm formation (Papapanou et al., 2018; Potempa et al., 2017; Teeuw et al., 2017). It is characterized by increased levels of subgingival pathogens, resulting in alveolar bone loss (ABL) and the progressive destruction of tooth-supporting structures (Ratheesh et al., 2018; Suvan et al., 2020). It is an important public health problem and is often called a silent disease because it rarely shows obvious signs and symptoms when it has progressed to its terminal stages (Papapanou & Susin, 2017). Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis are known as the major pathogens that induce periodontitis (Slots, 2017). Additionally, it is characterized by localized chronic inflammatory reactions with an increased serum cytokine level by pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, IL-10, and IL-17 (Ide et al., 2016; Potempa et al., 2017).

Periodontitis is treated through non-surgical and surgical treatments following the progress of the disease. In the case of non-advanced periodontitis, it is usually treated by nonsurgical methods, including scaling, root planing, and topical or oral antibiotic treatment (Costa et al., 2019; Matarese et al., 2017; Spanou et al., 2020). In contrast, patients with severe periodontitis required dental surgery such as bone grafting, flap surgery, and soft tissue grafting (Bai et al., 2018; Shrestha et al., 2018; Zufía et al., 2019). Therefore, many researchers have been developing new therapeutic approaches to investigate more effective therapy methods with bioactive substances for periodontitis and have reported that substances with antibacterial, antibiofilm, antimicrobial, anti-inflammatory, and anti-osteoclastic activities can be used as therapeutic agents for periodontitis (Kim et al., 2018; Shrestha et al., 2018; Spanou et al., 2020; Sun et al., 2019; Teughels et al., 2020).

Marine organisms, including Annelida, Chordata, Cnidaria, Phaeophyceae, and Rhodophyta, produce numerous bioactive substances since they have to face high competition for a unique living environment, such as high concentrations of salts, high pressure, and low concentration of oxygen (Batool et al., 2022; De Domenico et al., 2019; Jiang et al., 2012; Kim et al., 2016; Kim et al., 2022b; Oh et al., 2021). Additionally, many studies have investigated marine bioactive substances (MBS), including polyphenols, collagens (COL), polydeoxyribonucleotides, peptides, polysaccharides, chitins, and chitosans, and their therapeutic and pharmacological activities, such as antibacterial, antibiofilm, antimicrobial, antioxidant, and anti-inflammatory activities (Chandika et al., 2021; Heo et al., 2018; Kim et al., 2019a; Kim et al., 2020; Kim et al., 2021b; Kim et al., 2022a; Nguyen et al., 2016). For their huge biological activities, MBS has been increasing attention as a therapeutic and pharmaceutic agent for human diseases, including atopic dermatitis, idiopathic pulmonary fibrosis, osteoporosis, and periodontitis.

Based on these facts, this review summarizes the current state of MBS against periodontitis treatment, including the therapeutic effects and application forms of MBS. To facilitate their discussion, these breakthroughs will be classified as in vitro, in vivo, and clinical studies. Finally, we will discuss the challenges and recent trends in MBS research for periodontitis treatment.

Materials and Methods

Search strategy

We conducted a publication analysis on MBS against periodontitis with the use of the keywords “Marine” & “Periodontitis” via Google Scholar, PubMed, and Web of Science. We examined all language articles (2,580 articles) published from January 2019 to July 2022 (Fig. 1). In this research, we searched to find articles, which were conducted in vitro, in vivo (animal model), and clinical experiments, on the treatment of MBS against periodontitis. We reviewed the in vitro, in vivo, and clinical articles on the potential of MBS against periodontitis treatment throughout the article content (in the abstract, methods and materials, results, and discussion sections).

fas-26-1-1-g1
Fig. 1. Flowchart of the literation analysis.
Download Original Figure
Exclusion criteria

We excluded 1,496 several articles, such as symposium abstracts, reviews, book chapters, and dissertations, to avoid repetitive article counting. However, we examined the content of articles that were mentioned in the review articles and included them. Moreover, 1,035 studies simply described MBS or periodontitis in the introduction or discussion part. Therefore, we considered that 2,531 studies are irrelevant to the therapeutic effect and application forms of MBS against periodontitis and excluded them.

Inclusion strategy

Seven articles that conducted both in vitro and in vivo studies were counted as two. Thus, fifty-six studies were analyzed to investigate trends in the research and development of MBS against periodontitis treatment.

Results

Trend analysis by year of articles on marine bioactive substances (MBS) against periodontitis

Fig. 2 illustrates the number of studies on MBS for periodontitis published each year. We found that the number of studies on the therapeutic effect of MBS for periodontitis is increasing from 2019 to 2022. This trend indicated that characterizing the therapeutic efficacy and application of MBS, as a novel and safe treatment against periodontitis, is increasing interest.

fas-26-1-1-g2
Fig. 2. The number of articles about marine bioactive substances for periodontitis treatment from 2019 to 2022 (n = 56).
Download Original Figure
Trend analysis on extraction source of marine bioactive substances (MBS) against periodontitis

All studies (n = 56) were analyzed to identify the prevailing sources of MBS extraction (Fig. 3A) and twenty-three species, except unknown sources, were identified. Although extraction sources of most MBS did not know due to researchers purchasing MBS from material companies, MBS was mainly extracted from three classes (Phaeophyceae, Chordata, and Annelida) in the literature provided information on extraction sources.

fas-26-1-1-g3
Fig. 3. The number of articles about marine bioactive substances from 2019 to 2022 (n = 56). (A) Extraction source, (B) kind of study, and (C) treatment form.
Download Original Figure

Especially, Phaeophyceae (Brown algae) includes numerous genera, such as Ecklonia, Agarum, Sargassum, and Turbinaria, and contains potential bioactive substances, because it produces secondary metabolites, including alginic acid, fucoidan, polyphenol, alkaloids, and fatty acids (Tapia-Martinez et al., 2019). MBS extracted from Phaeophycea show antibacterial, anticoagulant, antithrombotic, antitumor, antiviral, and anti-inflammatory properties (Achmad et al., 2020b; Leal et al., 2018; Ren et al., 2019). Additionally, several biopolymers and bioactive substances, such as chitosan, COL, and fish oil, are extracted from Chordata and exhibit antioxidant, antimicrobial, and anti-inflammatory activities and biocompatibility (Asari et al., 2021; Oh et al., 2021; Satpathy et al., 2021). Arenicola marina inhabits intertidal soft sediments from the Arctic to the Mediterranean and contains peptides and extracellular hemoglobin with antimicrobial and anti-hepatotoxicity activities (De Cubber et al., 2019; Le Daré et al., 2021; Umnyakova et al., 2018). Based on the pharmacological properties of the marine resources, many studies analyzed in this review investigated the potential of MBS for periodontitis treatment.

Trend analysis of the potential effect of marine bioactive substances (MBS) against periodontitis

As mentioned in the method part, the experiments discussed herein will be classified as in vitro studies, in vivo studies, and clinical studies. Most experiments were conducted using in vitro models (31 studies), followed closely by in vivo studies (17 studies), suggesting that clinical studies (8 studies) are rarely conducted (Fig. 3B).

Trend analysis of marine bioactive substances (MBS) against periodontitis based on in vitro studies

Two studies have developed jellyfish COL-based scaffolds (Arslan & Kantarcioğlu, 2019; Rachmawati et al., 2021). One of these studies, a 2019 study fabricated Salvadora persica aqueous extract-loaded Rhizostoma pulmo COL (JC) scaffold and evaluated their effect on human periodontal ligament fibroblasts (HPLF) (Arslan & Kantarcioğlu, 2019). All fabricated scaffolds showed no cytotoxic effect and, in particular, the S. persica extract-unloaded JC scaffold enhanced cell proliferation 14 days after incubation. Other studies revealed that Aurelia aurita-derived COL scaffolds did not show cytotoxicity up to 1,000 μg/mL on human mesenchymal stem cells (hMSC) (Rachmawati et al., 2021).

Previous studies have confirmed that bioactive substances extracted from Phaeophyceae have therapeutic potential against periodontitis (Han et al., 2021; Jung et al., 2021; Kim et al., 2019b; Kim et al., 2021a; Magesh et al., 2020; Alden et al., 2022). Two of these studies investigated whether Ecklonia cava ethanol (EtOH) extract (ECE) has a therapeutic effect on periodontitis (Jung et al., 2021; Kim et al., 2019b). ECE presented no cytotoxicity in P. gingivalis lipopolysaccharide (Pg LPS)-stimulated RAW 264.7 and inhibited Pg LPS-induced production of inflammatory mediators, including nitric oxide (NO) and prostaglandin E2 (PGE2), and cyclooxygenase-2 (COX-2) activity (Kim et al., 2019b). Additionally, ECE decreased mRNA expression of inducible nitric oxygen synthase (iNOS), TNF-α, IL-1β, and IL-6 by activating the Nrf-2/heme oxygenase-1 (HO-1) pathway and suppressing the phosphorylation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway. The other study confirmed that ECE inhibited the production of PGE2 and reactive oxygen species (ROS) without cytotoxicity on human gingival fibroblast (HGF) (Jung et al., 2021). Another study investigated that Agarum clathratum, which has antioxidant and anti-microbial properties, exhibits potential for periodontitis treatment on Pg LPS-stimulated HGF (Han et al., 2021). A. clathratum methanol (MeOH) extract inhibited mRNA and protein expression of IL-1β without cytotoxicity on Pg LPS-stimulated HGF. A 2021 study isolated epiloliolide (ELL), a monoterpene lactone derived from the degradation of carotenoids, from Sargassum horneri and demonstrated that ELL promotes cell proliferation and migration in Pg LPS-stimulated human periodontal-derived ligament cell (HPDL) (Kim et al., 2021a). Additionally, ELL showed anti-inflammatory activity by suppressing protein expression of COX-2, iNOS, and NOD-like receptor pyrin domain-containing protein 3 (NLRP3) and mRNA expression and release of TNF-α, IL-1β, and IL-6. ELL induced cell mineralization and enhanced mRNA expression of alkaline phosphatase (ALP), osteopontin (OPN), and runt-related transcription factor 2 (RUNX-2) by activating the phosphorylation of the Protein kinase A (PKA)/cAMP response element-binding protein (CREB) pathway. Another study confirmed Turbinaria conoides petroleum ether (PE) extract has antibacterial activity on Streptococcus sp. and Actinomycetes sp. (Alden et al., 2022). The other study evaluated the antibacterial effect of Sargassum wightii PE extract on Streptococcus mutans (Magesh et al., 2020).

Rhodophyta is a groups of seaweeds with pharmacological properties and contains numerous natural compounds (Cotas et al., 2020). Therefore, two studies have investigated that extract and/or compounds from Rhodophyta have therapeutic potential for periodontitis (Cherian et al., 2019; Sharan & Vennila, 2021). A study obtained Kappaphycus alvarezii MeOH extract (KAM) and isolated marine bromophenol from KAM (KAB) (Cherian et al., 2019). KAB and KAM showed antibacterial activity on P. gingivalis and decreased Arginine specific gingipain (Rgp) activity and hemagglutination. Additionally, KAB and KAM suppressed the mRNA expression of peptidyl arginine deiminase (PPAD), Rgp, and hemagglutinin A (HgA). The other study obtained K. alvarezii extracts using different polarity solvents, including EtOH (KAE), ethyl acetate (KAEA), and chloroform (KAC) (Sharan & Vennila, 2021). After that, the authors evaluated the pharmacological effect of K. alvarezii extracts. All extracts showed high antioxidant activity and antimicrobial activity on Bacillus subtilis, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Candida albicans. Additionally, KAC showed high compatibility on Vero (Normal fibroblast cell line derived from African green monkey) and KAE decreased cell viability of KB-3-1 (Human epidermoid oral carcinoma cell line), which is an oral cancer cell line.

A 2020 study investigated the pharmacological activity of several EtOH extracts of Phaeophyceae and Rhodophyta on periodontitis (Yoon et al., 2020). In this study, EtOH extracts were obtained from 3 Phaeophyceae (Distromium decumbens [DDEE], Myelophycus simplex [MSEE], and Scytosiphon canaliculatus [SCEE]) and 3 Rhodophyta (Asparagopsis taxiformis [ATEE], Galaxaura fastigiata [GFEE], and Hypnea japonicay [HJEE]) and then evaluated their anti-inflammatory effect on Pg LPS-stimulated HGF. All extracts did not show cytotoxicity on Pg LPS-stimulated HGF. GFEE and SCEE suppressed only NO production, however, ATEE and HJEE inhibited the protein expression of iNOS, as well as NO production.

Three studies developed Chordata-derived biomaterial-based applications for periodontitis treatment (Achmad et al., 2020a; Elango et al., 2020; Zhou et al., 2020). One study confirmed whether Chanoschanos scale chitosan-based gel has high antibacterial activity on A. actinomycetemcomitans and P. gingivalis, similar to a positive control (Metronidazole) (Achmad et al., 2020a). A 2020 study has developed a tilapia COL/sodium alginate/titanium oxide (TiO2) 3D matrix (CB3D) (Elang et al., 2020). CB3D enhanced the level of ALP and osteocalcin (OCN) without cytotoxicity on HPLF. Additionally, CB3D increased the mRNA expression of COL, ALP, OCN, and RUNX-2 and protein expression of COL, OCN, and RUNX-2. These results indicate that CB3D promoted cell mineralization in HPLF. The other study developed a hydrogel based on tilapia COL and poly (vinyl alcohol) (PVA) (Zhou et al., 2020). When the increasing ratio of tilapia COL in hydrogel, hydrogel enhanced the cell viability, proliferation, and adhesion in HPLF and HGF. A 2020 study has fabricated tilapia COL/chitosan film, loaded with resveratrol or celastrol, as a dental implant for periodontitis (Wang et al., 2020b). Tilapia COL/chitosan film regardless of cross-linking using N-ethyl-N’-(3-(dimethylamino)propyl)carbodiimide/N-hydroxysuccinimide shows the highest cell proliferation and promotes the differentiation of bone marrow-derived macrophage (BMM) to tartrate-resistant acid phosphatase (TRAP)-positive cell.

Marine sponges belonging to the genus Agelas constitute several marine pyrrole-imidazole alkaloids with various biological properties (Rane et al., 2014). Therefore, one study investigated whether alkaloids isolated from the Agelas dilatata has therapeutic potential for periodontitis (Pech-Puch et al., 2020). In this study, eight alkaloids were isolated from A. dilatata and evaluated for antibacterial activity on P. aeruginosa. Bromoageliferin, an alkaloid isolated A. dilatata, has the highest antibacterial effect on P. aeruginosa (minimum inhibitory concentration = 8 μg/mL) and inhibited biofilm formation by P. aeruginosa.

A study confirmed that biseokeaniamide A, isolated Okeania sp., exhibits anti-inflammatory activity on LPS-stimulated RAW 264.7 (Ohno et al., 2020). Biseokeaniamide A inhibited NO production and IL-1β expression in LPS-stimulated RAW 264.7. Moreover, Biseokeaniamide A decreased protein expression iNOS by suppressing the phosphorylation of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα).

A 2020 study purified a cold-adapted dextranase (Cadex2870) from Catenovulum sp. and evaluated its biological activity for periodontitis treatment (Deng et al., 2020). Cadex2870 showed anti-biofilm activity on S. mutans and scavenging activity on the formed plaque biofilm of S. mutans.

HEMARINA-M101 (M101) is natural extracellular hemoglobin derived from the A. marina and is considered an extracellular oxygen carrier having high oxygen binding capacity (Batool et al., 2020). For these desirable properties of M101, M101 is used in anti-inflammatory and anti-fibrotic applications (Batool et al., 2020; Batool et al., 2022; Özçelik et al., 2021). One study investigated whether M101 has antibacterial and anti-inflammatory activity using Pg LPS-stimulated and P. gingivalis-infected human oral epithelial cell (HOEC) (Batool et al., 2020). M101 showed antibacterial activity on P. gingivalis. Additionally, M101 inhibited mRNA and protein expression of TNF-α and suppressed protein expression of IL-1β, IL-8, regulated upon activation, normal T cell expressed and presumably secreted (RANTES), and interferon gamma-induced protein (IP)-10 without cytotoxicity on inflammatory reaction-induced HOEC. Additionally, M101 inhibited mRNA expression of NF-κB and receptor activator of nuclear factors κB ligand (RANKL), enhanced the release of tissue inhibitors of matrix metalloproteinases (TIMP)-2, macrophage colony-stimulating factor human (M-CSF), and intercellular adhesion molecule 1 (ICAM-1), mRNA expression of Resolvin-E1 receptor, and protein expression of platelet-derived growth factor-BB (PDGF-BB), transforming growth factor (TGF)-β1, IL-10, IL-2, IL-4, IL-11, and IL-15. Moreover, another study investigated that M101 has therapeutic potential against P. gingivalis-infected or hypoxia-induced HOEC (Batool et al., 2022). M101 showed antioxidant and anti-hypoxic activities. Additionally, M101 increased cell migration and suppressed mRNA expression of glucose transporters (GLUT-1), GLUT-3, hypoxia-inducible factor (HIF)-1α, and matrix metalloproteinase (MMP)-9. To apply M101 for periodontitis treatment, a 2021 study fabricated hyaluronic acid-based hydrogel containing M101 (Xn2%-HA1%-M101) (Özçelik et al., 2021). The fabricated hydrogel showed antibacterial activity on P. gingivalis without cytotoxicity on HOEC.

One study investigated whether liposomes extracted from the skin of Dosidicus gigas have a potential for periodontitis treatment (Kao et al., 2021). In the RANKL/M-CSF-stimulated RAW 264.7 model, squid skin liposomes show an anti-inflammatory effect by suppressing the PGE2 level. Additionally, the squid skin inhibited osteoclast maturation by inhibiting TRAP activity and mRNA expression of NFATc1, c-Src, receptor activator of nuclear factor κB (RANK), Cathepsin K, TRAP, and dendritic cell-specific transmembrane protein (DC-STAMP).

Nine studies did not indicate information on extraction sources due to purchasing from material companies (Campana et al., 2020; Guo et al., 2021; Johnson et al., 2020; Li et al., 2021; Özdoğan et al., 2021; Ribeiro-Vidal et al., 2020; Tang et al., 2022; Xu et al., 2020; Zhou et al., 2021). One of these studies fabricated doxycycline-containing chitosan/carboxymethyl chitosan nanoparticles (Dox:CS/CMCS-NPs) and evaluated several biological activities, including antibacterial, anti-inflammatory, and cell proliferative activities (Xu et al., 2020). The fabricated nanoparticles exhibited an antibacterial effect on P. gingivalis and suppressed mRNA and protein expression of IL-1β without cytotoxicity on HGF stimulated by a combination treatment of Pg LPS and adenosine triphosphate (ATP). Additionally, Dox:CS/CMCS-NPs inhibited mRNA and protein expression of NLRP3 and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and mRNA expression and activation of Caspase-1. A 2020 study investigated that surfactin-loaded ĸ-carrageenan oligosaccharide-entangled cellulose nanofibers have therapeutic potential against periodontitis (Johnson et al., 2020). The fabricated nanofibers showed compatibility on RAW 264.7 and exhibited antibacterial activity on P. gingivalis and anti-biofilm activities on P. gingivalis and S. mutans. Moreover, nanofibers suppressed the metabolic activity of P. gingivalis and S. mutans, enhanced the malondialdehyde (MDA) level, and induced cell death of P. gingivalis and S. mutans. Another study on the biological activity of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) demonstrated that EPA and DHA have antibacterial activity on Actinomyces naeslundii, Fusobacterium nucleatum, Streptococcus oralis, Veillonella parvula, P. gingivalis, and A. actinomycetemcomitans and anti-biofilm activity on Streptococcus sp. and Aggregatibacter sp. (Ribeiro-Vidal et al., 2020). Another study also investigated the therapeutic effect of EPA using Pg LPS (normal and heat-inactivated form)-stimulated HGF (Zhou et al., 2021). In this study, they confirmed that EPA has an anti-inflammatory effect by suppressing mRNA expression of IL-1β and IL-6 and protein expression of IL-6 on Pg LPS-stimulated HGF. Moreover, EPA inhibited mRNA and protein expression of HO-1.

A 2020 study demonstrated that 2,2-bis(6-bromo-1H-indol-3-yl) ethanamine and its fluorinated analog, one of the marine bisindole alkaloids and its analog, have antibacterial, antimicrobial, and anti-biofilm activities on S. mutans. Additionally, both compounds showed a viable but non-culturable cell (VBNC) inducing effect and decreased oxacillin resistance against S. mutans. Another study developed gel formulations of atorvastatin solid dispersions with various mixture ratios (chitosan, lactic acid, pluronic F-68, and polyethylene glycols) (Özdoğan et al., 2021). All gel formations suppressed the release of IL-1β, IL-6, IL-8, IL-10, TGF-β1, TGF-β2, and TGF-β3. A 2021 study fabricated ginsenoside Rg1 and amelogenin-loaded injectable hydrogel, composed of glycol chitosan and aldehyde-modified hyaluronic acid, and evaluated its cytotoxicity (Guo et al., 2021). This study confirmed that the fabricated hydrogel did not show cytotoxicity on human bone marrow mesenchymal cells (HBMSC). Another 2021 study developed minocycline-loaded mouthwash containing natural polysaccharides, including chitosan and cyclodextrin (Li et al., 2021). The developed mouthwash showed the antibacterial and anti-biofilm effects on P. gingivalis. The other study investigated whether fucoidan-derived carbon dots (FDCD) have the biological potential for periodontitis treatment (Tang et al., 2022). FDCD showed antibacterial and anti-biofilm activity against Enterococcus faecalis and induced bacterial cell leakage. FDCD has non-cytotoxicity on MC3T3-E1 and RAW 264.7. Additionally, FDCD promoted ROS production in bacterial cells and increased macrophage migration.

Trend analysis of marine bioactive substances (MBS) against periodontitis based on in vivo studies

Stichopus herrmanni has pharmacological effects, such as antibacterial, antifungal, and wound healing effects (Sarhadizadeh et al., 2014; Wahyuningtyas et al., 2019). Based on these activities, three studies investigated the therapeutic potential of S. herrmanni for periodontitis (Adam et al., 2022; Mulawarmanti et al., 2019a; Mulawarmanti et al., 2019b). One of these studies confirmed using the femoral defect guinea pig model that S. herrmanni-extracted bone grafts induced OCN expression and increased the number of osteoblast at the implanted site (Adam et al., 2022). Others investigated that sodium carboxymethylcellulose (CMC-Na) gel containing S. herrmanni powder has a therapeutic activity on periodontitis diabetes rat model and periodontitis mice, respectively (Mulawarmanti et al., 2019a; Mulawarmanti et al., 2019b). In a periodontitis diabetes rat model induced by P. gingivalis administration, fabricated CMC-Na gel inhibited the expression of IL-1β and IL-10 (Mulawarmanti et al., 2019a). In a periodontitis mouse model induced by P. gingivalis administration, fabricated gel promoted osteoprotegerin (OPG) expression. Additionally, the fabricated gel increased osteoblast number and decreased osteoclast number at the alveolar mandible (Mulawarmanti et al., 2019b).

One study extracted sulfated polysaccharides from Gracilaria caudate, one of Rhodophyta, and evaluated their preventive effect on tissue damage caused by periodontitis (da Silva et al., 2019). G. caudate-derived sulfated polysaccharides suppressed gingival bleeding index (GBI), probing pocket depth (PPD), and myeloperoxidase (MPO) activity in gingiva and decreased MDA concentration and glutathione (GSH) levels in the liver in a ligature-induced periodontitis rat model. Additionally, sulfated polysaccharides prevent the histopathological alteration, which is induced by periodontitis.

Two studies demonstrated that ECE exhibits a therapeutic effect on periodontitis (Jung et al., 2021; Kim et al., 2019b). In a ligature-induced periodontitis rat model, ECE decreased several clinical symptoms, including ABL, bone resorption, the distance between the cementoenamel junction (CEJ) and the crest of the alveolar bone (ABC), gingival index (GI), periodontal bone height, and tooth mobility (TM). Additionally, ECE inhibited the proliferation and erosion of periodontal epithelial cells and suppressed the mRNA expression of IL-1β, MMP-2, MMP-9, and RANKL/OPG.

A 2020 study has isolated bromoageliferin from A. dilatata and assessed its efficacy against periodontitis-related microbial, such as Acinetobacter baumannii, K. pneumoniae, and P. aeruginosa (Pech-Puch et al., 2020). Bromoageliferin showed an anti-survival effect on P. aeruginosa in the P. aeruginosa-infected Galleria mellonella model.

Two studies using M101 as a therapeutic agent for periodontitis have been conducted (Batool et al., 2020; Batool et al., 2022). In one of these studies, M101 decreased clinical abscess size, inflammatory cell infiltration, inflammatory score, and TRAP-positive cell number and promoted the quantity and quality of bone in a P. gingivalis-induced subcutaneous calvarial abscess mice model (Batool et al., 2022). In P. gingivalis soaked ligature-induced periodontitis mouse model, M101 showed augmentation in fibrous connective tissue attachment (FA) and suppressed epithelial attachment (EA) (Batool et al., 2020). Additionally, M101 enhanced the attachment level (AL) and bone contour and quality. In histological analysis, M101 suppressed the expression of HIF-1α, NF-κB, and MMP-9 and the number of TRAP-positive cells at the periodontitis-induced site.

A 2020 study investigated whether fish oil has the potential for periodontitis treatment in a hypercholesterolemic rat model with ligature-induced periodontitis (Antona et al., 2020). Fish oil prevents bone loss and promotes bone trabeculae, the substitution of saturated fat, periodontal bone support (PBS), and a percentage of lower bone volume (LBV) and total lower volume (TLV).

Nannochloropsis oculata, one of the green microalgae, has been explored for functional food, nutraceutical, pharmacological agents, and aquaculture feed application (Andriopoulos et al., 2022; Hussein et al., 2020; Samarakoon et al., 2013). Therefore, Revianti et al. investigated whether N. oculata extract shows a pharmacological effect on periodontitis using an A. actinomycetemcomitans-infected periodontitis rat model (Revianti et al., 2020). Oral irrigation with N. oculata extracts decreased TNF-α expression and enhanced expression of OPG and IL-10 in periodontal ligament tissue.

One study demonstrated that Spirulina maxima extract has the potential for periodontitis treatment (Kang et al., 2021). S. maxima extract suppressed MPO activity, decreased TRAP-positive cell number in the destroyed space of the bone surface, and shorten the distance between CEJ and ABC in P. gingivalis-inoculated periodontitis rat model. Additionally, S. maxima extract increased OPG/RANKL expression ratio and osteoblast number in the destroyed space of the bone surface and inhibited mRNA expression of pro-inflammatory cytokines (TNF-α, IL-1β, IL-4, IL-6, and NF-κB), MMP-8, and MMP-13.

Two studies assessed the therapeutic effect of omega-3 polyunsaturated fatty acids on periodontitis using a ligature-induced periodontitis rat model (Jalal et al., 2020a; Jalal et al., 2020b). Omega-3 polyunsaturated fatty acids inhibited serum levels of IL-1β, IL-6, and TNF-α, decreased inflammatory cell infiltration and periodontal damage score, and enhanced bone formation at incisor periodontal tissue.

Three studies used chitosan as a base material for drug delivery systems for periodontitis treatment (Gani et al., 2022; Guo et al., 2021; Li et al., 2021). Ginsenoside Rg1 is a component of the natural extract of ginseng and is reported to its anti-inflammatory activities (Gao et al., 2020). Amelogenin is the main component of enamel matrix proteins and exhibits a regenerative effect on periodontal tissue (Liao et al., 2020). To deliver two drugs (Ginsenoside Rg1 and amelogenin) to periodontal tissue, one study fabricated Ginsenoside Rg1 and amelogenin-loaded injectable hydrogel composed of glycol chitosan and aldehyde-modified hyaluronic acid (1% HA-CHO + 1% GC + FeCl3) and evaluated the periodontium regenerative activity using rat model induced by periodontium defect (Guo et al., 2021). 1% HA-CHO + 1% GC + FeCl3 hydrogels nearly completed periodontium regeneration in the periodontium defect site and decreased clinical symptoms, such as the distance between CEJ and ABC, bone mineral density (BMD), bone volume fraction, trabecular thickness (Tb. Th), infiltration of inflammatory cells, and optical density of IL-1, TNF-α, and TGF-β. Another study developed a chitosan/cyclodextrin drug delivery system loaded with minocycline, which can inhibit the biofilm formation of Streptococcus gordonii and P. gingivalis (Elnagdy et al., 2021; Li et al., 2021). The developed drug delivery system alleviated several histological changes, including GI, TM, the distance between CEG and ABC, proliferation and erosion of periodontal epithelial cells, and bone resorption. Additionally, the system suppressed the mRNA expression of IL-1β, MMP-2, MMP-9, and RANKL/OPG ratio. The other study investigated whether hydroxyapatite-loaded chitosan gel as a bone graft has a therapeutic effect on periodontitis in a femoral defect rat model (Gani et al., 2022). Chitosan gel enhanced Bone morphogenetic protein (BMP)-2 expression and inhibited IL-1 expression at defect sites.

Trend analysis of marine bioactive substances (MBS) against periodontitis based on clinical studies

Few clinical studies (n = 8) have revealed that several MBS can be prevent and treat periodontitis (Deshpande et al., 2021, dos Santos et al., 2020; Hussain et al., 2022; Kujur et al., 2020; Murgia & Aspriello, 2021; Poornima et al., 2021; Pușcașu et al., 2022; Stańdo et al., 2020).

It was conducted two studies that used Chordata as an extraction source of base material or pharmacological agents (Deshpande et al., 2021; Pușcașu et al., 2022). Forty patients with chronic periodontitis were examined to investigate the therapeutic effects of the grafts with nanocrystalline hydroxyapatite in the absence and presence of fish COL (Deshpande et al., 2021). Through clinical parameter analysis, they confirmed that both grafts reduced plaque index (PI), GI, PPD, clinical attachment level (CAL), and radiographic defect depth, but no statistically significant difference between the two groups was observed. Another study demonstrated the potential of marine fish extracts (Alflutop®), which is known as extracts of Engraulis encrassicholusponticus, Sprattus sprattus sprattus, and Rizeafca nordmanni, for periodontitis treatment (Pușcașu et al., 2022). 44 patients with periodontitis received Alflutop® treatment after scaling, root planning, and debridement (SRP) and 44 patients with periodontitis received only SRP. Alflutop® treatment after SRP decreased GI of the Score 2 patients and alleviated the papilla bleeding index (PBI) and pathological TM of the Score 1 patients without side effects. The degree of TM and probing depth (PD) of all patients was decreased by Alflutop® treatment.

Almost clinical studies have evaluated the therapeutic effect of commercial products, such as omega-3 fatty acids, chitosan-based varnish, chitosan brush, COL-loaded composite, and Alfalife (omega-3 fatty acids) (dos Santos et al., 2020; Hussain et al., 2022; Kujur et al., 2020; Murgia & Aspriello, 2021, Poornima et al., 2021; Stańdo et al., 2020).

Four studies investigated the therapeutic effect of omega-3 fatty acids on patients with periodontitis (dos Santos et al., 2020; Kujur et al., 2020; Murgia & Aspriello, 2021; Stańdo et al., 2020). The results of three studies indicated that omega-3 fatty acids showed no adverse events and no changes in systemic parameters, including body mass index, blood pressure, levels of glycated hemoglobin A1c and triglycerides, in patients with periodontitis (dos Santos et al., 2020; Kujur et al., 2020; Stańdo et al., 2020). Additionally, omega-3 fatty acids decreased several indexes, such as Bleeding on probing (BoP), CAL, GI, PD, PI, PPD, and gingival recession (REC), the concentration of inflammatory mediators (IL-1β, IL-6, IL-8, IL-17, and IFN-γ), and release of chemokine (C-C motif) ligand (CCL)21, CCL26, CCL27, chemokine (C-X-C motif) ligand (CXCL)10, and CXCL16. In contrast, the treatment of omega-3 fatty acids increased the concentration of anti-inflammatory cytokine (IL-10), fibroblast growth factor (FGF)-2, and granulocyte-colony stimulating factor (G-CSF) and enhanced the release of CC4, CCL5, CCL15, CCL25, and chemokine (C-X3-C motif) ligand (CX3CL) 1. Another study investigated that Alfalife, a products containing omega-3 and omega-6 dietary supplements, has an anti-periodontitis effect in patients with periodontitis (Murgia & Aspriello, 2021). Alfalife decreased BoP, CAL, and PD. Based on these results, omega-3 fatty acids have a therapeutic potential for periodontitis through an anti-inflammatory effect.

Chitosan is a natural alkaline polysaccharide obtained from chitin via deacetylation and has antibacterial, anti-cancer, and antiviral activities, biocompatibility, and biodegradability (Ke et al., 2021; Wang et al., 2020a). Therefore, many studies have used chitosan as base materials for biomedical applications, such as drug delivery systems, hydrogels, nanofibers, and scaffolds (Cui et al., 2021; Peers et al., 2020; Rajabi et al., 2021; Sadeghianmaryan et al., 2020). For these reasons, two studies investigated the therapeutic effect of chitosan brush in patients undergoing fixed orthodontic therapy and in periodontitis patients (Hussain et al., 2022; Poornima et al., 2021). In patients undergoing fixed orthodontic therapy, chitosan brush showed no adverse events and decreased plaque bacterial count (Poornima et al., 2021). In patients with stage III and IV, grade B periodontitis, patients underwent no adverse events and decreased bleeding on probing (BoP) and PPD (Hussain et al., 2022).

This review analyzed and summarized the contents of related studies (Tables 13).
Table 1. Effects of marine bioactive substances against periodontitis through in vitro studies
Publication year Kind of sample Sample Kind of source Organisms Stimulator Cell Outcomes Ref
2019 Application S. persica extract-loaded COL hybrid constructs Cnidaria Rhizostoma pulmo - HPLF ↑Cell proliferation Arslan & Kantarcioğlu, 2019
2019 Extract ECE Phaeophyceae Ecklonia cava Pg LPS RAW 264.7 Non cytotoxicity
↓ NO and PGE2 production
↓ COX-2 activity
↓ mRNA expression of IL-1β, IL-6, iNOS, and TNF-α
↑ Activation of Nrf-2/HO-1 pathway
↓ Phosphorylation of NF-κB pathway
Kim et al., 2019b
2019 Compound Marine bromophenol & extract Rhodophyta Kappaphycus alvarezii - - Antibacterial effect on Porphyromonas gingivalis
↓ Rgp activity
↓ Hemagglutination
↓ mRNA expression of HgA, PPAD, and Rgp
Cherian et al., 2019
2020 Application Chitosan-loaded gel Chordata Chanos chanos - - Antibacterial effect on P. gingivalis and Aggregatibacter actinomycetemcomitan Achmad et al., 2020a
2020 Application COL-sodium alginate-TiO2 matrix Chordata Tilapia - HPLF Non cytotoxicity
↑Level of ALP and OCN
↑Mineralization
↑mRNA expression of ALP, COL, OCN, and RUNX-2
↑Protein expression of COL, OCN, and RUNX-2
Elango et al., 2020
2020 Application Dox:CS/CMCS-NPs Unknown Unknown Pg LPS & ATP HGF Antibacterial effect on P. gingivalis
Non cytotoxicity
↓ mRNA and protein expression of ASC and NLRP3
↓ mRNA and protein expression of IL-1β
↓ mRNA expression and activation of Caspase-1
Xu et al., 2020
2020 Application PVA/COL hydrogel Chordata Tilapia - HPLF & HGF ↑Cell viability and proliferation when increased COL ratio in both cell lines
↑Cell adhesion when increased COL ratio in both cell lines
Zhou et al., 2020
2020 Application Resveratrol and Celastrol-loaded fish COL/chitosan film Combination
(Chordata & unknown)
Tilapia & unknown - HPLF & BMM ↑Proliferation of HPLF
↑Differentiation to TRAP-positive cell from BMM
Wang et al., 2020a
2020 Application Surfactin-loaded ĸ-carrageenan oligosaccharides entangled cellulose nanofibers Unknown Unknown - RAW 264.7 Non cytotoxicity
Antibacterial effect on P. gingivalis
Anti-biofilm effect of P. gingivalis and Streptococcus mutans
↓ Metabolic activity of P. gingivalis and S. mutans
↑MDA level on P. gingivalis and S. mutans
↑Cell death of P. gingivalis and S. mutans
Johnson et al., 2020
2020 Compound Alkaloids Porifera Agelas dilatata - - Antibacterial effect on P. aeruginosa
Anti-biofilm effect on P. aeruginosa
Pech-Puch et al., 2020
2020 Compound Biseokeaniamide A Cyanobacteria Okeania sp. LPS RAW 264.7 Non cytotoxicity
↓ NO production
↓ Expression of IL-1β
↓ Protein expression of iNOS
↓ Protein expression of IκBα
Ohno et al., 2020
2020 Compound Cadex2870 Pseudomonadota Catenovulum sp. - - Anti-biofilm effect on S. mutans
Scavenging effect on the formed plaque biofilm of S. mutans
Deng et al., 2020
2020 Compound DHA & EPA Unkonwn Unkonwn - - Antibacterial effect on Streptococcus oralis, Actinomyces naeslundii, Veillonella parvula, Fusobacterium nucleatum, P. gingivalis, and Aggregatibacter actinomycetemcomitans
Anti-biofilm effect on Streptococcus sp. and Aggregatibacter sp.
Ribeiro-Vidal et al., 2020
2020 Compound M101 Annelida Arenicola marina Pg LPS stimulation or P. gingivalis infection HOEC Antibacterial effect on P. gingivalis
Non cytotoxicity
↓ mRNA expression of NF-κB, TNF-α, and RANKL
↓ Protein expression of TNF-α
↓ Protein expression of IL-1β, IL-8, IP-10, and RANTES
↑Protein expression of IL-10, IL-2, IL-4, IL-11, IL-15, PDGF-BB, and TGF-β1
↑Release of ICAM-1, M-CSF, and TIMP-2
↑mRNA expression of Resolvin-E1 receptor
Batool et al., 2020
2020 Compound 2,2-bis(6-bromo-1H-indol-3-yl) ethanamine & its fluorinated analogue Unknown Unknown - - Antimicrobial activity on S. mutans
Antibacterial activity against S. mutans in plasma
Anti-biofilm effect against S. mutans
Inducted VBNC
↓ Oxacillin resistance in S. mutans
Campana et al., 2020
2020 Extract Algae EtOH extracts
(ATEE, DDEE, GFEE, HJEE, MSEE, & SCEE)
Rhodophyta & Phaeophyceae ATEE, DDEE, GFEE, HJEE, MSEE & SCEE Pg LPS HGF Non cytotoxicity
↓ NO production
(ATEE, HJEE, GFEE, and SCEE)
↓ Protein expression of iNOS
(ATEE and HJEE)
Yoon et al., 2020
2021 Application Atorvastatin solid-loaded gel formulation Unknown Unknown TNF-α HGF ↓ Release of IL-1β, IL-6, IL-8, IL-10, TGF-β1, TGF-β2, and TGF-β3 Özdoğan et al., 2021
2021 Application Ginsenoside Rg1 and amelogenin-loaded injectable glycol chitosan/aldehyde-modified hyaluronic acid hydrogel Unknown Unknown - HBMSC Non cytotoxicity Guo et al., 2021
2021 Application Minocycline-loaded chitosan/cyclodextrin mouthwash Unknown Unknown - - Antibacterial effect on P. gingivalis Anti-biofilm effect on P. gingivalis Li et al., 2021
2021 Application Xn2%-HA1%-M101 gels Annelida A. marina - HOEC Non cytotoxicity
Antibacterial effect on P. gingivalis
Özçelik et al., 2021
2021 Extract Agarum clathratum MeOH extract Phaeophyceae A. clathratum Pg LPS THP-1 Non cytotoxicity
↓ mRNA and protein expression of IL-1β
Han et al., 2021
2021 Extract ECE Phaeophyceae E. cava LPS HGF Non cytotoxicity
↓ PGE2 production
↓ ROS production
↓ Phosphorylation of MAPK pathway
↓ mRNA expression of COX-2, IL-6, MMP-2, and MMP-8
↓ mRNA expression of MIP-1α and SDF-1
Jung et al., 2021
2021 Extract KAE, KAEA, KAC Rhodophyta K. alvarezii - Vero & KB-3-1 High antioxidant activity
Antimicrobial activity on Bacillus subtilis, Escherichia coli, Klebsiella pneumonia, Pseudomonas aeruginosa and Candida albicans
High cell viability on Vero (KAC)
High cytotoxicity on KB-3-1 (KAE)
Sharan & Vennila, 2021
2021 Extract Squid skin liposome Mollusca Dosidicus gigas RANKL/M-CSF RAW 264.7 Non cytotoxicity
↓ PGE2 level
↓ Osteoclast maturation
↓ Bone resorption pit area
↓ TRAP activity
↓ mRNA expression of NFATc1, c-Src, Cathepsin K, DC-STAMP RANK, and TRAP
Kao et al., 2021
2021 Compound EPA Unknown Unknown Pg LPS or heat-inactivated Pg LPS HGF Non cytotoxicity
↓ mRNA expression of IL-1β and IL-6
↓ Protein expression of IL-6
↑mRNA and protein expression of HO-1
Ke-cong et al., 2021
2021 Compound Epiloliolide Phaeophyceae Sargassum horneri Pg LPS HPDL ↑Proliferation, migration, mineralization
↓ Protein expression of NLRP3, COX-2, and iNOS
↑Phosphorylation of PKA/CREB pathway
↑mRNA expression of ALP, OPN, and RUNX-2
↓ mRNA expression of IL-1β, IL-6, and TNF-α
↓ Release of IL-1β, IL-6, and TNF-α
Kim et al., 2021a
2022 Application FDCD Unknown Unknown - MC3T3-E1 & RAW 264.7 Antibacterial effect on Enterococcus faecalis
Anti-biofilm effect on E. faecalis
Induction of cell leakage
Non cytotoxicity
↑ROS production inside bacterial cell
↑Macrophage migration
Tang et al., 2022
2022 Application Jellyfish COL scaffolds Cnidaria Aurelia aurita - hMSC ↑Cell proliferation Rachmawati et al., 2021
2022 Compound M101 Annelida A. marina P. gingivalis at MOI 100 or inducing hypoxia with CoCl2 HOEC Antioxidant effect
Anti-hypoxic effect
↑Cell migration
↓ mRNA expression of GLUT-1, GLUT-3, HIF-1α, and MMP-9
Batool et al., 2022
2022 Extract Turbinariaconoides PE extract Phaeophyceae T. conoides - - Antibacterial effect on Streptococcus sp. and Actinomycetes sp. Magesh et al., 2022
2022 Extract Sargassum Wightii PE extract Phaeophyceae S. Wightii - - Antibacterial effect on S. mutans Magesh et al., 2020

HPLF, human periodontal ligament fibroblast; ECE, Ecklonia cava ethanol extract; Pg LPS, Porphyromonas gingivalis lipopolysaccharide; NO, nitric oxide; PGE2, prostaglandin E2; COX-2, cyclooxygenase-2; IL, interleukin; iNOS, inducible nitric oxygen synthase; TNF-α, tumor necrosis factor alpha; HO-1, heme oxygenase-1; Rgp, Arginine specific gingipain; HgA, hemagglutinin A; PPAD, peptidyl arginine deiminase; ALP, alkaline phosphatase; OCN, osteocalcin; COL, collagen; RUNX-2, runt-related transcription factor 2; Dox:CS/CMCS-NPs, doxycycline-containing chitosan/carboxymethyl chitosan nanoparticles; ATP, adenosine triphosphate; HGF, human gingival fibroblast; ASC, apoptosis-associated speck-like protein containing a caspase recruitment domain; NLRP3, NOD-like receptor pyrin domain-containing protein 3; PVA, poly (vinyl alcohol); BMM, bone marrow-derived macrophages; TRAP, tartrate resistant acid phosphatase; MDA, malondialdehyde; IκBα, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha; DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; HOEC, human oral epithelial cells; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; RANKL, receptor activator of nuclear factors κB ligand; RANTES, regulated upon activation, normal T cell expressed and presumably secreted; PDGF-BB, platelet-derived growth factor-BB; TGF, transforming growth factor; ICAM-1, intercellular adhesion molecule 1; M-CSF, macrophage colony-stimulating factor; TIMP, tissue inhibitors of matrix metalloproteinases; VBNC, viable but non-culturable cells; EtOH, ethanol; ATEE, Asparagopsis taxiformis; DDEE, Distromium decumbens; GFEE, Galaxaura fastigiata; HJEE, Hypnea japonicay; MSEE, Myelophycus simplex; SCEE, Scytosiphon canaliculatus; HBMSC, human bone marrow mesenchymal cell; MeOH, methanol; THP-1, human monocytic leukemia cell; ROS, reactive oxygen species; MMP, matrix metalloproteinase; MIP-1α, macrophage inflammatory protein-1 alpha; SDF, stromal cell-derived factor; KAE, Kappaphycus alvarezii extracts ethanol extract; KAEA, Kappaphycus alvarezii extracts ethyl acetate extract; KAC, Kappaphycus alvarezii extracts chloroform extract; DC-STAMP, dendritic cell specific transmembrane protein; HPDL, human periodontal derived ligament cells; PKA, protein kinase A; CREB, cAMP response element-binding protein; OPN, osteopontin; FDCD, fucoidan-derived carbon dots; hMSC, human mesenchymal stem cell; GLUT, glucose transporters; HIF, hypoxia inducible factor; PE, petroleum ether.

Download Excel Table
Table 2. Effects of marine bioactive substances against periodontitis through in vivo studies
Publication year Kind of sample Sample Kind of source Organisms Model Outcomes Ref
2019 Application CMC-Na gel containing Stichopus herrmanni powder Echinodermata S. herrmanni Periodontitis diabetes rat model induced by Porphyromonas gingivalis administration ↓ Expression of IL-1β and IL-10 Mulawarmanti et al., 2019a
2019 Application CMC-Na gel containing S. herrmanni powder Echinodermata S. herrmanni Periodontitis mice model induced by P. gingivalis administration ↑OPG expression
↑Number of osteoblast
↓ Number of osteoclast
Mulawarmanti et al., 2019b
2019 Extract Sulfated polysaccharides Rhodophyta Gracilaria caudata Ligature-induced periodontitis rat model ↓ GBI and PPD
↓ MPO activity
↓ MDA concentration and GSH level in liver
↓ Histopathological alteration in liver tissue
da Silva et al., 2019
2019 Extract ECE Phaeophyceae Ecklonia cava Ligature-induced periodontitis rat model ↓ GI and TM
↓ Distance between CEJ and ABC
↓ Proliferation and erosion of periodontal epithelial cell
↓ Bone resorption
↓ mRNA expression of IL-1β, MMP-2, MMP-9, and RANKL/OPG
Kim et al., 2019b
2020 Compound Bromoageliferin Porifera Agelas dilatata Pseudomonas aeruginosa-infected Galleria mellonella model Anti-survival effect Pech-Puch et al., 2020
2020 Compound M101 Annelida Arenicola marina P. gingivalis-induced subcutaneous calvarial abscess mice model ↓ Clinical abscess size
↓ Inflammatory cell infiltration
↓ Inflammatory score
↑Quantity and quality of bone
↓ TRAP-positive cell number
Batool et al., 2020
2020 Compound Omega-3 polyunsaturated fatty acid Unknown Unknown Ligature-induced periodontitis rat model ↓ Serum level of IL-1β, IL-6, and TNF-α Jalal et al., 2020a
2020 Compound Omega -3 polyunsaturated fatty acid Unknown Unknown Ligature-induced periodontitis rat model ↑Bone formation
↓ Inflammatory cell infiltration
↓ Periodontal damage score
Jalal et al., 2020b
2020 Extract Fish oil Chordata Unknown Hypercholesterolemic rat model with ligature-induced periodontitis ↓ Bone loss
↑Bone trabeculae
↑LBV/TLV%
↑Substitution of saturated fat
↑PBS
Antona et al., 2020
2020 Extract Nannochloropsis oculata extract Ochrophyta N. oculata A. actinomycetemcomitans infected periodontitis rat model ↓ TNF-α expression
↑Expression of OPG and IL-10
Revianti et al., 2020
2021 Application Ginsenoside Rg1 and amelogenin-loaded injectable glycol chitosan/aldehyde-modified hyaluronic acid hydrogel Unknown Unknown Periodontitis rat model induced by periodontium defect Nearly completed periodontium regeneration
↓ Distance between CEJ and ABC
↓ BMD, bone volume fraction (BV/TV), and Tb. Th
↓ Infiltration of inflammatory cells
↓ Optical density of IL-1, TGF-β, and TNF-α
Guo et al., 2021
2021 Application Minocycline-loaded chitosan/cyclodextrin mouthwash Unknown Unknown Ligature-induced periodontitis rat model Antiplaque biofilm activity
↓ GBI and ABL
↓ Distance between CEJ and ABC
Li et al., 2021
2021 Extract ECE Phaeophyceae E. cava Ligature-induced periodontitis rat model ↓ ABL and bone height
↓ mRNA expression of RANKL/OPG
Jung et al., 2021
2021 Extract Spirulina maxima extract Cyanophyta S. maxima P. gingivalis inoculated periodontitis rat model ↓ MPO activity
↓ TRAP-positive cell number
↑OPG/RANKL expression ratio
↓ Distance between CEJ and ABC
↑Osteoblast number
↓ mRNA expression of IL-1β, IL-4, IL-6, NF-κB, and TNF-α
↓ mRNA expression of MMP-8 and MMP-13
↑Protein expression OPN, OSX, and Type I COL
↑Activation of BMP-2/Smad pathway
Kang et al., 2021
2022 Application Hydroxyapatite-loaded chitosan gel Unknown Unknown Femoral defect rat model ↑BMP-2 expression
↓ IL-1 expression
Gani et al., 2022
2022 Compound M101 Annelida A. marina P. gingivalis soaked ligature-induced periodontitis mice model Augmentation in the FA
↓ EA
↑AL and bone contour and quality
↓ Expression of HIF-1α, NF-κB, and MMP-9
↓ Number of TRAP positive cell
Batool et al., 2022
2022 Extract S. herrmanni extract Echinodermata S. herrmanni Femoral defect guinea pig model ↑OCN expression
↑Number of osteoblast
Adam et al., 2022

CMC-Na, sodium carboxymethylcellulose; OPG, osteoprotegerin; ECE, Ecklonia cava ethanol extract; GBI, gingival bleeding index; PPD, probing pocket depth; MPO, myeloperoxidase; MDA, malondialdehyde; GSH, glutathione; GI, gingival index; TM, tooth mobility; CEJ, cementoenamel junction; ABC, the crest of the alveolar bone; IL, interleukin; MMP, matrix metalloproteinase; RANKL, receptor activator of nuclear factors κB ligand; TRAP, tartrate resistant acid phosphatase; TNF-α, tumor necrosis factor alpha; LBV, lower bone volume; TLV, total lower volume; PBS, periodontal bone support; BMD, bone mineral density; BV, bone volume; TV, tissue volume; Tb. Th, trabecular thickness; TGF, transforming growth factor; ABL, alveolar bone loss; OPN, osteopontin; OSX, osterix; COL, collagen; BMP, bone morphogenetic protein; FA, fibrous connective tissue attachment; EA, epithelial attachment; AL, attachment level; HIF, hypoxia inducible factor; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells.

Download Excel Table
Table 3. Effects of marine bioactive substances against periodontitis through clinical studies
Publication year Kind of sample Sample Kind of source Organisms Patient Outcomes Ref
2020 Compound Omega-3 fatty acids Unknown Unknown Periodontitis patient
(n = 1, female)
No changes in systemic parameters
↓ BoP, CAL, PD, and PI
↓ Concentration of IFN-γ, IL-1β, and IL-6 in gingival crevicular fluid
dos Santos et al., 2020
2020 Compound Omega-3 fatty acids Unknown Unknown Periodontitis patients
(n = 30, both)
No adverse events
↓ BoP, CAL, PD, and REC
↓ Concentration of IL-8 and IL-17
↑Concentration of IL-10
↑Release of CCL4, CCL5, CCL15, CCL25 and CX3CL1
↓ Release of CCL21, CCL26, CCL27, CXCL1, CXCL10 and CXCL16
↑Concentration of FGF2 and G-CSF
Stańdo et al., 2020
2020 Compound Omega-3 fatty acids Unknown Unknown Patients with chronic generalised moderate periodontitis (n = 76, both) ↓ PPD, CAL, and GI Kujur et al., 2020
2021 Application Chitosan brush Unknown Unknown Patients undergoing fixed orthodontic therapy (n = 20, both) No adverse events
↓ Plaque bacterial count
Poornima et al., 2021
2021 Application COL-loaded composite Chordata Unknown Periodontitis patients (n = 40, both) ↓ PI, GI, PPD, and CAL
↓ Radiographic defect depth
Deshpande et al., 2021
2021 Compound Alfalife Unknown Unknown Periodontitis patients (n = 24, both) ↓ BoP, CAL, and PD Murgia & Aspriello, 2021
2022 Application Chitosan brush Unknown Unknown Patients with stage III and IV, grade B periodontitis (n = 78, both) No adverse event
↓ BoP and PPD
Hussain et al., 2022
2022 Extract Alflutop® treatment after SRP Chordata Engraulis encrassicholus ponticus, Sprattus sprattus sprattus, & Rizeafca nordmanni Periodontitis patients (n = 88, both) No side effects
↓ GI of the score 2 patients
↓ PBI of the score 1 patients
↓ Pathological TM of the score 1 patients
↓ Degree of TM and PD
Pușcașu et al., 2022

BoP, bleeding on probing; CAL, clinical attachment level; PD, probing depth; PI, plaque index; IL, interleukin; REC, gingival recession; CCL, chemokine (C-C motif) ligand; CX3CL, chemokine (C-X3-C motif) ligand; CXCL, chemokine (C-X-C motif) ligand; FGF, fibroblast growth factor; G-CSF, granulocyte-colony stimulating factor; PPD, probing pocket depth; GI, gingival index; SRP, scaling, root planning, and debridement; PBI, papilla bleeding index; TM, tooth mobility.

Download Excel Table

Discussion and Conclusion

All studies (56 articles), which were published from January 2019 to July 2022 were analyzed and summarized literature content about the therapeutic effects and application forms of MBS for periodontitis.

Analysis result of the published year indicated that publication numbers steadily is increasing from January 2019. Most studies used commercial MBS obtained from material companies. However, as illustrated in Fig. 3A, MBS against periodontitis treatment, which in described information of marine organisms, was mainly extracted from Phaeophyceae, Chordata, and Annelida. Additionally, MBS was investigated in the form of extracts, compounds, and applications (e.g., brush, film, hydrogel, mouthwash, nanofiber, and nanoparticle) at a similar percentage during the analyzed period (Fig. 3C).

Biomedical engineering is the application of engineering principles and problem-solving techniques to medical and biological research for healthcare that provides new strategies for the diagnosis, prevention, and treatment of various human diseases (Kim & Jung, 2019; Kim et al., 2021a; Oh et al., 2021). The main goal of biomedical engineering is the development of new therapies and technologies to help, recover, and replace damaged tissue and organs without side effects (Chandika et al., 2021; Lee et al., 2020; Oh et al., 2020). Although numerous biomedical applications incorporating MBS have been investigated, most studies investigated the regenerative effect of MBS on skin and bone (Chandika et al., 2021; Kim & Jung, 2019; Kim et al., 2021a; Lee et al., 2020; Oh et al., 2020; Oh et al., 2021). In this review, only 18 studies developed several biomedical applications, including film, hydrogel, nanofiber, and nanoparticles (Achmad et al., 2020a; Arslan & Kantarcioğlu, 2019; Deshpande et al., 2021; Elango et al., 2020; Gani et al., 2022; Guo et al., 2021; Hussain et al., 2022; Johnson et al., 2020; Mulawarmanti et al., 2019a; Mulawarmanti et al., 2019b; Özçelik et al., 2021; Özdoğan et al., 2021; Poornima et al., 2021; Rachmawati et al., 2021; Tang et al., 2022; Wang et al., 2020a; Xu et al., 2020; Zhou et al., 2020). The biomedical application for periodontitis is steadily investigated from 2019 to 2022. Therefore, we suggest that biomedical engineering incorporating MBS for periodontitis has a high potential as a therapeutic strategy.

Although MBS has high therapeutic potential on periodontitis, several limitations and weaknesses remain in terms of extraction source, underlying mechanism from in vitro and in vivo, and few results in clinical safety and efficacy. Firstly, although the ocean covers more than 70% of the earth embedding numerous marine organisms with biological agents, only a few marine organisms were used as extraction sources of bioactive substances for periodontitis treatment (Kim et al., 2019a). Next, most studies used commercial products of MBS, not extracted MBS. Thus, it is hard to confirm the specific information of extraction sources and active compounds. The underlying mechanisms from in vitro and in vivo of MBS as therapeutic agents for periodontitis were demonstrated by only seven articles (12.5% of the number of articles about MBS for periodontitis treatment) (Batool et al., 2020; Batool et al., 2022; Kang et al., 2021; Kao et al., 2021; Kim et al., 2019b; Kim et al., 2021a; Ohno et al., 2020). Finally, few clinical studies (8 articles) were conducted to confirm the safety and therapeutic efficacy of MBS.

Therefore, additional research is needed to develop more numerous extraction sources such as starfish, algae, and unstudied marine organisms, and isolate single compounds to apply medical devices. Clinical studies on the safety and therapeutic efficacy of MBS for periodontitis treatment are needed to demonstrate the therapeutic potential and safety for periodontitis patients. Finally, biomedical applications incorporating MBS for periodontitis must be constantly developed to enhance the therapeutic efficacy of MBS for periodontitis.

Competing interests

No potential conflict of interest relevant to this article was reported.

Funding sources

This research was financially supported by the Research Grant of Pukyong National University (2021).

Acknowledgements

Not applicable.

Availability of data and materials

Upon reasonable request, the datasets of this study can be available from the corresponding author.

Ethics approval and consent to participate

This article does not require IRB/IACUC approval because there are no human and animal participants.

References

1.

Achmad H, Djais AI, Jannah M, Carmelita AB, Uinarni H, Arifin EM, et al. Antibacterial chitosan of milkfish scales (Chanos chanos) on bacteria Prophyromonas gingivalis & Agregatibacter actinomycetemcomitans. Syst Rev Pharm. 2020a; 11:836-41.

2.

Achmad H, Huldani , Carmelita AB, Fauziah , Hidayah N, Bokov D. Antioxidant and antiviral potential of brown algae (Phaeophyceae). Int J Pharm Res. 2020b; 3:2117-25.

3.

Adam M, Achmad H, Nasir M, Putri SW, Azizah A, Satya DE. Stimulation of osteoblast and osteocalcin in the bone regeneration by giving bonegraft golden sea cucumber. J Int Dent Med Res. 2022; 15:140-7.

4.

Alden Schnyder Jason D, Magesh KT, Aravindhan R, Sathya Kumar M. Evaluation of antibacterial activity of marine brown algae Turbinaria conoides against oral pathogens. Res J Pharm Technol. 2022; 15:2236-9

5.

Andriopoulos V, Gkioni MD, Koutra E, Mastropetros SG, Lamari FN, Hatziantoniou S, et al. Total phenolic content, biomass composition, and antioxidant activity of selected marine microalgal species with potential as aquaculture feed. Antioxidants. 2022; 11:1320

6.

Antona ME, Ramos C, Stranges A, Monteiro AF, Chaves MMG, Mandalunis P, et al. Fish oil diet effects on alveolar bone loss, in hypercholesterolemic rats. Arch Oral Biol. 2020; 109:104553

7.

Arslan YE, Kantarcioğlu İ. Salvadora persica extract-laden jellyfish collagen hybrid constructs for periodontal tissue regeneration. J Turk Chem Soc Chem. 2019; 6:51-62

8.

Asari MA, Sirajudeen KNS, Yusof NAM, Amin MSIM. DHA-rich fish oil and Tualang honey reduce chronic stress-induced oxidative damage in the brain of rat model. J Tradit Complement Med. 2021; 12:361-6

9.

Bai YL, Zheng TJ, Zhang ZW, Gan Y, Huang J. Efficacy of minocycline hydrochloride combined with flap surgery for chronic periodontitis: a meta-analysis. West China J Stomatol. 2018; 36:421-7.

10.

Batool F, Petit C, Stutz C, Özçelik H, Gegout PY, Benkirane-Jessel N, et al. M101, a therapeutic oxygen carrier derived from Arenicola marina, decreased Porphyromonas gingivalis-induced hypoxia and improved periodontal healing. J Periodontol. 2022; 93:1712-24

11.

Batool F, Stutz C, Petit C, Benkirane-Jessel N, Delpy E, Zal F, et al. A therapeutic oxygen carrier isolated from Arenicola marina decreased P. gingivalis induced inflammation and tissue destruction. Sci Rep. 2020; 10:14745

12.

Campana R, Mangiaterra G, Tiboni M, Frangipani E, Biavasco F, Lucarini S, et al. A fluorinated analogue of marine bisindole alkaloid 2,2-bis(6-bromo-1H-indol-3-yl) ethanamine as potential anti-biofilm agent and antibiotic adjuvant against Staphylococcus aureus. Pharmaceuticals. 2020; 13:210

13.

Chandika P, Oh GW, Heo SY, Kim SC, Kim TH, Kim MS, et al. Electrospun porous bilayer nano-fibrous fish collagen/PCL bio-composite scaffolds with covalently cross-linked chitooligosaccharides for full-thickness wound-healing applications. Mater Sci Eng C. 2021; 121:111871

14.

Cherian C, Jannet Vennila J, Sharan L. Marine bromophenols as an effective inhibitor of virulent proteins (peptidyl arginine deiminase, gingipain R and hemagglutinin A) in Porphyromas gingivalis. Arch Oral Biol. 2019; 100:119-28

15.

Costa FO, Lages EJP, Lages EMB, Cota LOM. Periodontitis in individuals with liver cirrhosis: a case–control study. J Clin Periodontol. 2019; 46:991-8

16.

Cotas J, Leandro A, Pacheco D, Gonçalves AMM, Pereira L. A comprehensive review of the nutraceutical and therapeutic applications of red seaweeds (Rhodophyta). Life. 2020; 10:19

17.

Cui C, Sun S, Wu S, Chen S, Ma J, Zhou F. Electrospun chitosan nanofibers for wound healing application. Eng Regen. 2021; 2:82-90

18.

da Silva FRP, Pinto MSC, de Carvalho França LF, Alves EHP, dos Santos Carvalho J, Di Lenardo D, et al. Sulfated polysaccharides from the marine algae Gracilaria caudata prevent tissue damage caused by ligature-induced periodontitis. Int J Biol Macromol. 2019; 132:1-8

19.

De Cubber L, Lefebvre S, Lancelot T, Denis L, Gaudron SM. Annelid polychaetes experience metabolic acceleration as other Lophotrochozoans: inferences on the life cycle of Arenicola marina with a dynamic energy budget model. Ecol Model. 2019; 411:108773

20.

De Domenico S, De Rinaldis G, Paulmery M, Piraino S, Leone A. Barrel jellyfish (Rhizostoma pulmo) as source of antioxidant peptides. Mar Drugs. 2019; 17:134

21.

Deng T, Feng Y, Xu L, Tian X, Lai X, Lyu M, et al. Expression, purification and characterization of a cold-adapted dextranase from marine bacteria and its ability to remove dental plaque. Protein Expr Purif. 2020; 174:105678

22.

Deshpande AP, Baburaj MD, Tambe LV, Prasad U. Extracellular matrix containing nanocomposite bone graft in periodontal regeneration – a randomized controlled clinical and radiographic evaluation. J Indian Soc Periodontol. 2021; 25:313-9

23.

dos Santos NC, Araujo CF, Andere NMRB, Miguel MMV, Westphal MRA, Van Dyke T, et al. Omega-3 fatty acids and low-dose aspirin in the treatment of periodontitis and metabolic syndrome: case report. J Int Acad Periodontol. 2020; 22:223-30.

24.

Eke PI, Borgnakke WS, Genco RJ. Recent epidemiologic trends in periodontitis in the USA. Periodontol 2000. 2020; 82:257-67

25.

Elango J, Selvaganapathy PR, Lazzari G, Bao B, Wenhui W. Biomimetic collagen-sodium alginate-titanium oxide (TiO2) 3D matrix supports differentiated periodontal ligament fibroblasts growth for periodontal tissue regeneration. Int J Biol Macromol. 2020; 163:9-18

26.

Elnagdy S, Raptopoulos M, Kormas I, Pedercini A, Wolff LF. Local oral delivery agents with anti-biofilm properties for the treatment of periodontitis and peri-implantitis. A narrative review. Molecules. 2021; 26:5661

27.

Gani A, Yulianty R, Supiaty S, Rusdy M, Dwipa Asri G, Eka Satya D, et al. Effectiveness of combination of chitosan gel and hydroxyapatite from crabs shells (Portunus pelagicus) waste as bonegraft on periodontal network regeneration through IL-1 and BMP-2 analysis. Int J Biomater. 2022; 2022:1817236

28.

Gao H, Kang N, Hu C, Zhang Z, Xu Q, Liu Y, et al. Ginsenoside Rb1 exerts anti-inflammatory effects in vitro and in vivo by modulating toll-like receptor 4 dimerization and NF-kB/MAPKs signaling pathways. Phytomedicine. 2020; 69:153197

29.

Guo H, Huang S, Yang X, Wu J, Kirk TB, Xu J, et al. Injectable and self-healing hydrogels with double-dynamic bond tunable mechanical, gel–sol transition and drug delivery properties for promoting periodontium regeneration in periodontitis. ACS Appl Mater Interfaces. 2021; 13:61638-52

30.

Han JM, Cho SD, Chung GH, Choi SJ, Shin JA, Hong SD, et al. Agarum clathratum inhibits LPS-induced IL-1β expression through the regulation of the JNK/c-Jun signaling pathway. Korean J Oral Maxillofac Pathol. 2021; 45:111-20

31.

Heo SY, Ko SC, Nam SY, Oh J, Kim YM, Kim JI, et al. Fish bone peptide promotes osteogenic differentiation of MC3T3-E1 pre-osteoblasts through upregulation of MAPKs and Smad pathways activated BMP-2 receptor. Cell Biochem Funct. 2018; 36:137-46

32.

Hussain B, Karaca EO, Kuru BE, Gursoy H, Haugen HJ, Wohlfahrt JC. Treatment of residual pockets using an oscillating chitosan device versus regular curettes alone—a randomized, feasibility parallel-arm clinical trial. J Periodontol. 2022; 93:780-9

33.

Hussein HA, Mohamad H, Ghazaly MM, Laith AA, Abdullah MA. Anticancer and antioxidant activities of Nannochloropsis oculata and Chlorella sp. extracts in co-application with silver nanoparticle. J King Saud Univ Sci. 2020; 32:3486-94

34.

Ide M, Harris M, Stevens A, Sussams R, Hopkins V, Culliford D, et al. Periodontitis and cognitive decline in Alzheimer’s disease. PLOS ONE. 2016; 11e0151081

35.

Jalal AA, Al-Qassab ZA, Al-Rawi RA. Effects of the systemic administration of omega-3-polyunsaturated fatty acid on experimental periodontitis: a histopathological study for rat’s periodontal tissue. Ann Trop Med Public Health. 2020a; 23:SP231226

36.

Jalal AA, Al-Qassab ZA, Al-Rawi RA. Effects of the systemic administration of omega-3 polyunsaturated fatty acid on experimental periodontitis (immunological analysis for rat’s serum). Ann Trop Med Public Health. 2020b; 23:SP2311271

37.

Jiang CS, Müller WEG, Schröder HC, Guo YW. Disulfide- and multisulfide-containing metabolites from marine organisms. Chem Rev. 2012; 112:2179-207

38.

Johnson A, He JL, Kong F, Huang YC, Thomas S, Lin HTV, et al. Surfactin-loaded ĸ-carrageenan oligosaccharides entangled cellulose nanofibers as a versatile vehicle against periodontal pathogens. Int J Nanomedicine. 2020; 15:4021-47

39.

Jung JI, Kim S, Baek SM, Choi SI, Kim GH, Imm JY. Ecklonia cava extract exerts anti-inflammatory effect in human gingival fibroblasts and chronic periodontitis animal model by suppression of pro-inflammatory cytokines and chemokines. Foods. 2021; 10:1656

40.

Kang MS, Moon JH, Park SC, Jang YP, Choung SY. Spirulina maxima reduces inflammation and alveolar bone loss in Porphyromonas gingivalis-induced periodontitis. Phytomedicine. 2021; 81:153420

41.

Kao YF, Tu MC, Chai HJ, Lin YL, Chen YC. Suppressive effects of an apoptotic mimicry prepared from jumbo-flying squid-skin phospholipids on the osteoclastogenesis in receptor activator of nuclear factor kappa B ligand/macrophage colony-stimulating factor-induced RAW 264.7 cells. J Chin Med Assoc. 2021; 84:51-60

42.

Ke CL, Deng FS, Chuang CY, Lin CH. Antimicrobial actions and applications of chitosan. Polymers. 2021; 13:904

43.

Kim EN, Nabende WY, Jeong H, Hahn D, Jeong GS. The marine-derived natural product epiloliolide isolated from Sargassum horneri regulates NLRP3 via PKA/CREB, promoting proliferation and anti-inflammatory effects of human periodontal ligament cells. Mar Drugs. 2021a; 19:388

44.

Kim GH, Park HH, Chandika P, Ko SC, Jung KM, Yoon SC, et al. Chemical compositions and biological activities of marine invertebrates from the East Sea of South Korea. Fish Aquat Sci. 2019a; 22:13

45.

Kim H, Kim MB, Kim C, Hwang JK. Inhibitory effects of panduratin A on periodontitis-induced inflammation and osteoclastogenesis through inhibition of MAPK pathways in vitro. J Microbiol Biotechnol. 2018; 28:190-8

46.

Kim S, Choi SI, Kim GH, Imm JY. Anti-inflammatory effect of Ecklonia cava extract on Porphyromonas gingivalis lipopolysaccharide-stimulated macrophages and a periodontitis rat model. Nutrients. 2019b; 11:1143

47.

Kim SC, Heo SY, Oh GW, Yi M, Jung WK. A 3D-printed polycaprolactone/marine collagen scaffold reinforced with carbonated hydroxyapatite from fish bones for bone regeneration. Mar Drugs. 2022a; 20:344

48.

Kim TH, Heo SY, Oh GW, Heo SJ, Jung WK. Applications of marine organism-derived polydeoxyribonucleotide: its potential in biomedical engineering. Mar Drugs. 2021b; 19:296

49.

Kim TH, Heo SY, Oh GW, Park WS, Choi IW, Kang HW, et al. A phlorotannins-loaded homogeneous acellular matrix film modulates post-implantation inflammatory responses. J Tissue Eng Regen Med. 2022b; 16:51-62

50.

Kim TH, Jung WK. R&D trends of brown algae as potential candidates in biomedical application. J Mar Biosci Biotechnol. 2019; 11:1-13.

51.

Kim TH, Ko SC, Oh GW, Park HH, Lee DS, Yim MJ, et al. Studies on bioactive substances and antioxidant activities of marine algae from Jeju island. J Mar Biosci Biotechnol. 2016; 8:30-8

52.

Kim TH, Lee HS, Oh SJ, Hwang CW, Jung WK. Phlorotannins ameliorate extracellular matrix production in human vocal fold fibroblasts and prevent vocal fold fibrosis via aerosol inhalation in a laser-induced fibrosis model. J Tissue Eng Regen Med. 2020; 14:1918-28

53.

Kujur SK, Goswami V, Nikunj AM, Singh G, Bandhe S, Ghritlahre H. Efficacy of omega 3 fatty acid as an adjunct in the management of chronic periodontitis: a randomized controlled trial. Indian J Dent Res. 2020; 31:220-35

54.

Le Daré B, Ferron PJ, Bellamri N, Ribault C, Delpy E, Zal F, et al. A therapeutic oxygen carrier isolated from Arenicola marina decreases amanitin-induced hepatotoxicity. Toxicon. 2021; 200:87-91

55.

Leal D, Mansilla A, Matsuhiro B, Moncada-Basualto M, Lapier M, Maya JD, et al. Chemical structure and biological properties of sulfated fucan from the sequential extraction of subAntarctic Lessonia sp (Phaeophyceae). Carbohydr Polym. 2018; 199:304-13

56.

Lee HS, Jeong MS, Ko SC, Heo SY, Kang HW, Kim SW, et al. Fabrication and biological activity of polycaprolactone/phlorotannin endotracheal tube to prevent tracheal stenosis: an in vitro and in vivo study. J Biomed Mater Res Part B. 2020; 108:1046-56

57.

Li B, Shi L, Liu R, Li Z, Cao S, Li J. A lingering mouthwash with sustained antibiotic release and biofilm eradication for periodontitis. J Mater Chem B. 2021; 9:8694-707

58.

Liao Y, Li H, Shu R, Chen H, Zhao L, Song Z, et al. Mesoporous hydroxyapatite/chitosan loaded with recombinant-human amelogenin could enhance antibacterial effect and promote periodontal regeneration. Front Cell Infect Microbiol. 2020; 10:180

59.

Magesh KT, Aravindhan R, Kumar MS, Sivachandran A. Antibacterial efficacy of the extract of Sargassum Wightii against oral pathogen: an in vitro study. J Orofac Sci. 2020; 12:96-100.

60.

Matarese G, Ramaglia L, Cicciù M, Cordasco G, Isola G. The effects of diode laser therapy as an adjunct to scaling and root planning in the treatment of aggressive periodontitis: a 1-year randomized controlled clinical trial. Photomed Laser Surg. 2017; 35:702-9

61.

Mulawarmanti D, Parisihni K. Widyastuti. The effect of Sticopus hermanii-hyperbaric oxygen therapy to inflammatory response of diabetic periodontitis. IOP Conf Ser Earth Environ Sci. 2019a; 217; :012060

62.

Mulawarmanti D, Parisihni K, Widyastuti W. The effect of Sticopus hermaniii-hyperbaric oxygen therapy to osteogenesis of diabetic periodontitis. Diabetes Complicat. 2019b; 3:1-3

63.

Murgia MS, Aspriello SD. Dietary supplementation with omega-3 alpha-linoleic acid and polyunsaturated fatty acids (PUFAs): a new therapeutic protocol additional to scaling and root planning in the treatment of chronic periodontitis: a 6 months follow up. J Dent Oral Disord Ther. 2021; 9:1-7

64.

Nguyen VT, Ko SC, Oh GW, Heo SY, Jeon YJ, Park W, et al. Anti-inflammatory effects of sodium alginate/gelatine porous scaffolds merged with fucoidan in murine microglial BV2 cells. Int J Biol Macromol. 2016; 93:1620-32

65.

Oh GW, Nam SY, Heo SJ, Kang DH, Jung WK. Characterization of ionic cross-linked composite foams with different blend ratios of alginate/pectin on the synergistic effects for wound dressing application. Int J Biol Macromol. 2020; 156:1565-73

66.

Oh GW, Nguyen VT, Heo SY, Ko SC, Kim CS, Park WS, et al. 3D PCL/fish collagen composite scaffolds incorporating osteogenic abalone protein hydrolysates for bone regeneration application: in vitro and in vivo studies. J Biomater Sci Polym Ed. 2021; 32:355-71

67.

Ohno O, Terasaki T, Sano T, Hitomi Y, Miyamoto J, Matsuno K. Inhibitory effects of biseokeaniamide A against lipopolysaccharide-induced signal transduction. Bioorg Med Chem Lett. 2020; 30:127069

68.

Özçelik H, Batool F, Corre M, Garlaschelli A, Conzatti G, Stutz C, et al. Characterization of a hyaluronic acid-based hydrogel containing an extracellular oxygen carrier (M101) for periodontitis treatment: an in vitro study. Int J Pharm. 2021; 605:120810

69.

Özdoğan AI, Akca G, Şenel S. Development and in vitro evaluation of gel formulation of atorvastatin solid dispersions. J Drug Deliv Sci Technol. 2021; 61:102199

70.

Papapanou PN, Sanz M, Buduneli N, Dietrich T, Feres M, Fine DH, et al. Periodontitis: consensus report of workgroup 2 of the 2017 World Workshop on the Classification of Periodontal and Peri-Implant Diseases and Conditions. J Periodontol. 2018; 89:S173-82

71.

Papapanou PN, Susin C. Periodontitis epidemiology: is periodontitis under-recognized, over-diagnosed, or both?. Periodontol 2000. 2017; 75:45-51

72.

Pech-Puch D, Pérez-Povedano M, Martinez-Guitian M, Lasarte-Monterrubio C, Vázquez-Ucha JC, Bou G, et al. In vitro and in vivo assessment of the efficacy of bromoageliferin, an alkaloid isolated from the sponge Agelas dilatata, against Pseudomonas aeruginosa. Mar Drugs. 2020; 18:326

73.

Peers S, Montembault A, Ladavière C. Chitosan hydrogels for sustained drug delivery. J Control Release. 2020; 326:150-63

74.

Poornima P, Krithikadatta J, Ponraj RR, Velmurugan N, Kishen A. Biofilm formation following chitosan-based varnish or chlorhexidine-fluoride varnish application in patients undergoing fixed orthodontic treatment: a double blinded randomised controlled trial. BMC Oral Health. 2021; 21:465

75.

Potempa J, Mydel P, Koziel J. The case for periodontitis in the pathogenesis of rheumatoid arthritis. Nat Rev Rheumatol. 2017; 13:606-20

76.

Pușcașu CG, Caraiane A, Dumea E, Șachir EE, Raftu G, Pușcașu RA, et al. Measurement of the clinical effects of a marine fish extract on periodontal healing—a preliminary clinical interventional study. Appl Sci. 2022; 12:677

77.

Rachmawati R, Hidayat M, Permatasari N, Widyarti S. Jellyfish (Aurelia aurita) collagen scaffolds potential in alveolar bone regeneration. F1000research. 2021; 10:318.

78.

Rajabi M, McConnell M, Cabral J, Ali MA. Chitosan hydrogels in 3D printing for biomedical applications. Carbohydr Polym. 2021; 260:117768

79.

Rane R, Sahu N, Shah C, Karpoormath R. Marine bromopyrrole alkaloids: synthesis and diverse medicinal applications. Curr Top Med Chem. 2014; 14:253-73

80.

Ratheesh V, Subramanian S, Prakash PSG, Victor DJ. Evaluation of association of vitamin D receptor genetic polymorphism with severe chronic periodontitis in an Ethnic Tamilian population. Genet Test Mol Biomark. 2018; 22:615-21

81.

Ren D, Wang Q, Yang Y, Hu Y, Song Y, He Y, et al. Hypolipidemic effects of fucoidan fractions from Saccharina sculpera (Laminariales, Phaeophyceae). Int J Biol Macromol. 2019; 140:188-95

82.

Revianti S, Andriani D, Parisihni K, Wahjuningsih E. Effectiveness of oral irrigation with an extract of green microalga Nannochloropsis oculata as an anti-inflammatory in rats infected with Aggregatibacter actinomycetemcomitans. Biodivers J Biol Divers. 2020; 21:2977-81

83.

Ribeiro-Vidal H, Sánchez MC, Alonso-Español A, Figuero E, Ciudad MJ, Collado L, et al. Antimicrobial activity of EPA and DHA against oral pathogenic bacteria using an in vitro multi-species subgingival biofilm model. Nutrients. 2020; 12:2812

84.

Sadeghianmaryan A, Naghieh S, Sardroud HA, Yazdanpanah Z, Soltani YA, Sernaglia J, et al. Extrusion-based printing of chitosan scaffolds and their in vitro characterization for cartilage tissue engineering. Int J Biol Macromol. 2020; 164:3179-92

85.

Samarakoon KW, Nam KO, Ko JY, Lee JH, Kang MC, Kim D, et al. Purification and identification of novel angiotensin-I converting enzyme (ACE) inhibitory peptides from cultured marine microalgae (Nannochloropsis oculata) protein hydrolysate. J Appl Phycol. 2013; 25:1595-606

86.

Sarhadizadeh N, Afkhami M, Ehsanpour M. Evaluation bioactivity of a sea cucumber, Stichopus hermanni from Persian Gulf. Eur J Exp Biol. 2014; 4:254-8.

87.

Satpathy AA, Dash S, Das SK, Shyamasuta S, Pradhan S. Functional and bioactive properties of chitosan from Indian major carp scale. Aquacult Int. 2021; 29:417-30

88.

Sharan LV, Vennila JJ. Phyto-pharmacological investigation of marine red algae Kappaphycus alvarezii (Doty) doty ex silva for oral diseases. Int J Algae. 2021; 23:183-209

89.

Shrestha S, Shrestha SM, Pradhan A, Aryal S. Platelet rich fibrin and bone graft in the treatment of intrabony defect in periodontitis patients. J Nepal Soc Periodontol Oral Implantol. 2018; 2:49-55

90.

Slots J. Periodontitis: facts, fallacies and the future. Periodontol 2000. 2017; 75:7-23

91.

Spanou A, Nelson K, Ermer MA, Steybe D, Poxleitner P, Voss PJ. Primary wound closure and perioperative antibiotic therapy for prevention of bisphosphonate-related osteonecrosis of the jaw after tooth extraction. Quintessence Int. 2020; 51:220-8.

92.

Stańdo M, Piatek P, Namiecinska M, Lewkowicz P, Lewkowicz N. Omega-3 polyunsaturated fatty acids EPA and DHA as an adjunct to non-surgical treatment of periodontitis: a randomized clinical trial. Nutrients. 2020; 12:2614

93.

Sun X, Wang L, Lynch CD, Sun X, Li X, Qi M, et al. Nanoparticles having amphiphilic silane containing Chlorin e6 with strong anti-biofilm activity against periodontitis-related pathogens. J Dent. 2019; 81:70-84

94.

Suvan J, Leira Y, Moreno Sancho FM, Graziani F, Derks J, Tomasi C. Subgingival instrumentation for treatment of periodontitis A systematic review. J Clin Periodontol. 2020; 47:155-75

95.

Tang S, Zhang H, Mei L, Dou K, Jiang Y, Sun Z, et al. Fucoidan-derived carbon dots against Enterococcus faecalis biofilm and infected dentinal tubules for the treatment of persistent endodontic infections. J Nanobiotechnology. 2022; 20:321

96.

Tapia-Martinez J, Hernández-Cruz K, Franco-Colín M, Mateo-Cid LE, Mendoza-Gonzalez C, Blas-Valdivia V, et al. Safety evaluation and antiobesogenic effect of Sargassum liebmannii J. Agardh (Fucales: Phaeophyceae) in rodents. J Appl Phycol. 2019; 31:2597-607

97.

Teeuw WJ, Kosho MXF, Poland DCW, Gerdes VEA, Loos BG. Periodontitis as a possible early sign of diabetes mellitus. BMJ Open Diabetes Res Care. 2017; 5e000326

98.

Teughels W, Feres M, Oud V, Martín C, Matesanz P, Herrera D. Adjunctive effect of systemic antimicrobials in periodontitis therapy: a systematic review and meta-analysis. J Clin Periodontol. 2020; 47:257-81

99.

Umnyakova ES, Gorbunov NP, Zhakhov AV, Krenev IA, Ovchinnikova TV, Kokryakov VN, et al. Modulation of human complement system by antimicrobial peptide arenicin-1 from Arenicola marina. Mar Drugs. 2018; 16:480

100.

Wahyuningtyas E, Hsu LC, Lan WC, Wen SC, Ou KL, Chou HH, et al. Application of a promising bone graft substitute in bone tissue regeneration: characterization, biocompatibility, and in vivo animal study. BioMed Res Int. 2019; 2019:1614024

101.

Wang R, Bao B, Bao C, Wang S, Ur Rahman S, Hou C, et al. Resveratrol and celastrol loaded collagen dental implants regulate periodontal ligament fibroblast growth and osteoclastogenesis of bone marrow macrophages. Chem Biodivers. 2020b; 17e2000295

102.

Wang W, Meng Q, Li Q, Liu J, Zhou M, Jin Z, et al. Chitosan derivatives and their application in biomedicine. Int J Mol Sci. 2020a; 21:487

103.

Xu S, Zhou Q, Jiang Z, Wang Y, Yang K, Qiu X, et al. The effect of doxycycline-containing chitosan/carboxymethyl chitosan nanoparticles on NLRP3 inflammasome in periodontal disease. Carbohydr Polym. 2020; 237:116163

104.

Yoon HS, Kim HJ, Park CM. Anti-inflammatory activity of various seaweeds in LPS-PG stimulated HGF-1 cells. J Korean Soc Oral Health Sci. 2020; 8:25-30

105.

Zhou K, Sun M, Xia Y, Xie Y, Shu R. Effects of eicosapentaenoic acid on biological activity and inflammatory factor expression of human gingival fibroblasts. Shanghai J Stomatol. 2021; 30:225-31.

106.

Zhou T, Zheng K, Sui B, Boccaccini AR, Sun J. In vitro evaluation of poly (vinyl alcohol)/collagen blended hydrogels for regulating human periodontal ligament fibroblasts and gingival fibroblasts. Int J Biol Macromol. 2020; 163:1938-46

107.

Zufía J, Blasi G, Gómez-Meda R, Blasi A. The four-layer graft technique, a hard and soft tissue graft from the tuberosity in one piece. J Esthet Restor Dent. 2019; 31:304-10