An Overview of the Use of Medicinal Plants in Regenerative Dentistry

Abstract

Aim: The oral cavity has the particularity to host multiple hard and soft tissues, in this paper, we will discuss the current therapies that lead to cell differentiation by regenerative therapies and the future alternatives proposed by medicinal plants and all the regenerative potential of these different tissues. Material and Methods: A detailed review of the literature through the various search engines: Scopus, PubMed, google scholar, Cochrane, etc., uses the selected keywords to explore the effect of the regenerative potential of several medicinal plants. Results: Through our research, we have proceeded to sort different medicinal plants, according to their repairing and regenerative potential on the different tissues of the oral cavity. Conclusion: Future studies are conceivable to explore the opportunities and potential provided by medicinal plants in the field of regenerative dentistry.

Share and Cite:

Dhoum, S. , Ibenmoussa, S. and Sidqui, M. (2023) An Overview of the Use of Medicinal Plants in Regenerative Dentistry. Open Journal of Stomatology, 13, 50-88. doi: 10.4236/ojst.2023.131005.

1. Introduction

As defined by Mason and Dunnill, the subject of regenerative medicine aims to replace and regenerate human cells, tissues or organs [1].

Herbs and plants have been used to cure many diseases since ancient times, and their medicinal and therapeutic importance has been proven in the prevention and treatment of many pathologies.

Inexpensive and without major side effects, several studies have shown that natural compounds, especially secondary metabolites isolated from plants, have many biological and medicinal activities such as antibacterial, antiviral, antifungal, anticancer, analgesic, anti-inflammatory, regenerative and antitumoral.

The role of medicinal plants in disease treatment has been the subject of many studies in odontology, especially for their antibacterial and anti-inflammatory potential, their regenerative potential needs to be explored.

2. Particularities of Oral Cavity

The human teeth are composed of organized, mineralized tissue layers of dentin, cementum and enamel. In native tooth architecture, an enamel-encased crown surrounds the internal pulp chamber and roots [2].

Enamel is derived from oral epithelium tissue, while dentin, pulp and periodontium derive from the neural crest.

In healthy tooth anatomy, the dentin-pulp complex lies below a continuous layer of ordered enamel, protecting the vessel and nerve rich pulp [2].

In the dentin layer, odontoblasts create and regulate tissue matrix components. Epithelial-mesenchymal interactions are essential for the transition of mesenchymal embryonic pulp cells to the pre-odontoblastic stage. Signaling molecules from the inner enamel epithelium encourage differentiation of peripheral dental papilla cells, odontoblast precursors, which eventually become secondary odontoblasts [3] [4] [5].

Human dental pulp stem cells (DPSCs) originate from migrating neural crest cells, are derived from the embryonic ectoderm layer and possess mesenchymal stem cell properties. This feature confers them vast differentiation potential, in addition to their ability to secrete trophic factors and their immunoregulatory properties. DPSCs can differentiate into odontoblasts, osteocytes/osteoblasts, adipocytes, chondrocytes, or neural cells [6] [7].

DPSCs can also regenerate dental tissue composed of vascular, connective, and neural tissues. During tooth development, primitive ectomesenchyme becomes enclosed within the prospective teeth to form the dental pulp, a rich source of stem cells. Odontogenesis (the process of tooth development) involves the cell matrix of types and specific cellular processes which result in the differentiation, growth, maturation and eruption of developing teeth in the mouth [5] [6].

2.1. Soft Tissues

Gingiva and oral mucosa: The gingiva is the oral mucosa that covers the alveolar bone and cervical part of the tooth. It forms a physical barrier against oral bacteria and provides mechanical protection to the underlying tissues. Given its protective nature, the gingiva is a key for wound healing since is constantly exposed to trauma or bacterial products that lead to inflammatory and infectious events.

The gingiva is composed of a layer of epithelial tissue (divided into three functional compartments; gingival, sulcular, and junctional epithelium) and connective tissue: superficial and deep compartments [8].

The cellular processes of oral soft tissue are similar to the healing of skin wounds. Nevertheless, it is commonly stated that oral wounds heal better and with less scar formation than dermal wounds.

The normal response to injury involves three overlapping stages: Inflammation, new tissue formation and remodeling.

After tissue injury, distinct biological pathways immediately become activated and are synchronized to prevent infection and restore the damaged tissues.

The cells recruited during wound healing include components of the immune system, endothelial cells, keratinocytes, and fibroblasts [8] [9].

Pulp tissue: The dental pulp is a soft ecto-mesenchymal tissue surrounded by dentin and it is highly vascularized and highly innerved.

The pulp is composed of 75% water, 25% organic material, heterogeneous population of cells: fibroblastic cells, stem cells, capillary blood vessels, peripheral nerves, lymphatic elements, as well as cells from the immune system and an extracellular matrix: fibers, and fundamental substance, concentration of Collagen fibers to support blood vessels and nerves [10].

The pulp tissue is unique given its volume and its confinement within the dentin. It has low blood supply, except in the apical foramen, and lacks collateral blood supply.

Dental pulp is an important component of teeth and plays important roles: Formation of the dentin (odontoblasts), Nutrition: the pulp is responsible for nourishing the dentine through the odontoblastic extensions and metabolites coming from the pulp vascular system, the essential function of the pulp and it is maintained as long as the pulp is vital, Sensitivity: in the face of stimuli, aggression or pain and Defense/repair: ability to form dentin face to an aggression.

There is a strong belief that the dental pulp offers a potentially viable source of MSCs for regenerative medicine-based applications. Such principles are based on their availability and ease of isolation [4] [10].

2.2. Hard Tissues

Enamel: The main challenge in the regeneration of enamel is its acellular nature.

Dental enamel is a calcified tissue that forms the outer protective shield of the anatomical crown of a tooth, it’s produced by ameloblasts. These enamel forming cells go through apoptosis when amelogenesis is finalized and they are lost when the tooth erupts. The in vitro culture of ameloblasts is yet unestablished in a scale needed for appropriate tissue regeneration [3] [11].

Mature enamel is mainly composed of Inorganic material 96% hydroxyapatite crystals (HA), 3% of water and 1% proteins: amelogenin, tuftelin … [3].

Actual research follows a biomimetic approach by using amelogenin, peptide fragments of amelogenin, or various synthetic peptides as a template matrix to mimic the environment for the deposition of enamel [3].

Dentin: It constitutes the most mineralized part of the tooth, it is covered and protected by the enamel in the dental crown and by the cement in the radicular part.

The responsible cells in the dentin formation are the odontoblasts, these cells are long-living post-mitotic cells and they are not replaced throughout the whole life of the tooth.

Its inorganic matrix is composed of HA crystals while its organic matrix is mainly composed of a collagenous structural component, formed of collagen type I (about 90%), collagen type III and V, and a small quantity of organic matrix molecules (proteoglycans of chondroitin sulfate (biglycan and decorin), heparan sulfate (perlecan and entactin), keratan and dermatan sulfate. It has also non-collagen proteins that participate in the mineralization process [12].

These proteins are members of the SIBLING family (small integrin-binding ligand N-linked glycoprotein) and include the sialophosphoprotein (DSPP), Dentin Matrix Protein 1 (DMP-1), osteopontin (OPN), Matrix Extracellular Phosphoglycoprotein (MEPE), and Integrin-Binding Sialoprotein (IBSP). DSPP and DMP-1 are considered specific markers for these cells [13].

Dental Cementum: It is a hard and avascular connective tissue located on the dental root; the main function of cementum is to connect the fibers of the PDL that emerge from the alveolar bone to the root. It is involved in the development, repair, and regeneration of periodontal support structures [14] [15].

We can distinguish two types of cementum: Cellular and acellular.

Their composition is distributed:

· 50% of mineral: Hydroxyapatite Ca10(PO4)6(HO)2;

· 50% of organic matrix: type I collagen 90%, cement-related collagen protein includes type III collagen, Cementum Attachment Protein (HACD1/CAP), Cementum protein 1 (CEMP-1);

· Cells: predominance of fibroblasts, the epithelial cell rests of Malassez, monocytes, macrophages, cementoblasts, osteoblasts, myofibroblasts, nerve cells, epithelial cells, endothelial cells and a low level of stem cells [14] [15].

Alveolar Bone: It’s the portion of the bone that surrounds the teeth and represents the primary support structure for the teeth.

Two of the main causes of alveolar bone loss are periodontitis and trauma.

The procedures for the regeneration of the alveolar bone are mainly based on the use of natural or synthetic scaffolds and bioactive agents (Figure 1).

However, there are some inherent limitations associated with biomaterials and traditional techniques [16] [17].

Composition:

· Inorganic matrix 67%.: calcium, phosphate and calcium hydroxyapatite crystals.

· Organic matrix 33%: collagen and non-collagenous materials.

· Cells: osteoblasts, osteocytes and osteoclasts.

Figure 1. Overview of the applications of polyphenols in dental field [19].

3. Use of Medicinal Plants in the Oral Cavity

Regenerative, antibacterial, antiviral, anti-inflammatory and analgesic effects of medicinal plants on oral tissues have been proved by several studies, numerous active principals have been studied and have shown interesting results in terms of accelerating the healing process [18] [19].

3.1. Action on Enamel

Enamel remineralization includes the incorporation of minerals such as carbonate, magnesium, sodium, fluoride, calcium and phosphate, among others. Generally, a direct association is observed between these ions in saliva and reduced tooth enamel demineralization (dental caries) [20] [21].

The effect of natural compounds on enamel remineralization has already been proved and clinically used with the Casein phosphopeptide-amorphous calcium phosphate (CPP-ACP) complex [22].

The anticariogenic effect of several medicinal plants has been subject of many studies (Table 1): Galla chinesis that contains metabolites such as tannins and flavonoids, promoting the mineral apposition in cavity lesions [23] [24] [25] [26], Carboxymethyl chitosan, Ilex guayusa have being studied for their anticariogenic effect and enhancing enamel remineralization [27] [28] [29] [30], Piper marginatum have been studied for its antifungal properties but has shown cytotoxicity in an invivo study on rats [27] [29] [31] [32] [33], Prilla frutescens seed shown an antibacterial effect and preventing dental caries [26] [34], and Coconut fruit extracts: virgin coconut oil (VCO), coconut milk (CM), and coconut water (CW) hold properties which enable them to prevent dental caries. Previous

Table 1. Summary based on the literature review of the medicinal plants studied with a potential of enamel reparation, remineralization and protection.

studies found that VCO exhibited anti-microbial properties while CM contained high content of calcium [35] [36].

3.2. Action on Dentin-Pulp Complex

Regenerative endodontic treatment is defined as the biological-based processes that enable replacement of damaged structures of the dentin-pulp complex (Murray et al. 2007) [37]. The main purpose behind the regenerative endodontic treatment is to regain normal physiological features such as innate immunity, tertiary dentinogenesis, sense of occlusal pressure and pain that the necrotic pulp does not aloud.

Three elements are necessary to achieve pulpal regeneration: dental stem cells, biomaterials, and growth factors [38] [39].

Over the past decade, interest in drugs derived from medicinal plants has markedly increased, the commun known treatments are based on synthetic bioactive materials such: Biodentin®, Mineral Trioxyd Aggregate (MTA), Calcium hydroxide based materials [38] [39].

Some of medicinal plants have been studied for their regenerative potential since 1982 by llewicz et al (39), several studies (Table 2) have been conducted to explore the action of Propolis (apis mellifera) and Aloe vera (acemannan) that proved their clinical efficiency to induce odontogenic differentiation equal to synthetic products currently used for the same purpose [40] - [60]. Pinus massoniana have been studied [40] for inducing odontogenic differentiation, also Astragalus membnaceus [41], curcuma longa [42] [43] [44], Camellia sinensis [45] and Cinnamomum verum [43] [45], Allium sativum have shown an acceleration on the healing process and antibacterial effects [46].

Cafeic acid combined to MTA have shown angiogenic properties and formation of hydroxyapatite, immunosuppressive and odontogenic properties [47] [48] [49], Proanthocyanidins induces odontogenic differentiation and increases the expression of biomineralization and odontogenic differentiation regulators: RUNX2, BMP2, OCN and DSPP [36] [46] [47] [48] [40] [50] [51] [52].

Schisandra chinensis is effective in anti-oxidative mechanisms in dental pulp cells [53] [54] [55] [56].

3.3. Action on Cementum and Periodontal Ligament

The process of periodontal tissue regeneration is initiated at the moment that the damage takes place by the production of growth factors and cytokines by the damaged and inflammatory cells.

New therapeutic approaches to periodontal regeneration using molecular approaches have emerged over the last twenty years:

· Growth factors,

· Platelet-derived growth factor,

· Insulin-like growth factors,

· Transforming growth factor-beta1,

· Basic fibroblast growth factor,

· Dexamethasone Bone morphogenetic proteins.

Medicinal plants offer a new field of exploration of their regenerative potential, but still more studies are needed to evaluate their efficiency in cementum regeneration.

Icariin [82] [83] [84] proved their action in regulation of osteoclast differentiation and enhancement of cementum repair, Acemannan induces an acceleration of the healing process in periodontal disease [85] [86] [87] [88] [89], Baicalin induce periodontal tissue regeneration [90], Salvadora persica (siwak/miswak) [91] and Rutin (buckwheat, japanese pagoda tree) [92] showed their interest in maintaining periodontal health.

3.4. Action on Alveolar Bone

Periodontitis is commonly characterized by the formation of intrabony defects.

Table 2. Summary based on the literature review of the medicinal plants studied with an action on dentin-pulp complex.

Multiple surgical approaches for treating these defects have shown effectiveness in improving clinical and radiographic parameters. Moreover, histologic evidence demonstrates the potential to achieve regeneration of the periodontal attachment: new bone, cementum, and periodontal ligament (PDL) using different therapeutic approaches [93] [94].

The implication of medicinal plants in bone regeneration has been the subject of many studies and experiment (Table 3).

Berberine and Icariin promote odontoblastic and odontogenic differentiation, they are both responsible of the inhibition of alveolar bone osteoporosis and promote periodontal bone regeneration [43] [82] [95] - [102], Drynaria iisos increases bone cell viability [103] [104], Rhizoma drynariae enhances the proliferation of hmscs [2] [103] [104], Foeniculum vulgare promotes differentiation of hMSCs into osteoblasts [104], Ferula gummosa has antibacterial effects and induces proliferation of hMSCs into osteocytes [105], Resveratrol induces proliferation and differentiation of articular cartilage, enhances bone healing by its anti-inflammatory properties, responsible for the inhibition of boneloss and increases the blood supply [104] [106] - [111]. Fructus ligustri lucidi increases osteogenesis stimulating genes [112] [113].

Osthole (cnidium monnieri/angelica pubescens) has osteoprotective activity, osteogenic activity, and induces bone formation [114] [115] [116] [117] [118], China herba epimedii improves osteogenesis and inhibition of osteoclasts in hMSCs [119], Naringin (rhizoma drynariae) has been the subject of several studies that proved its effect on the proliferation and osteogenic differentiation of

Table 3. Summary based on the literature review of the medicinal plants studied with an action on cementum, periodontal ligament and alveolar bone.

hMSCc [120] [121], Ginkgo biloba promotes osteogenesis [122], Tanshinol and salvionic acid b (salvia miltiorrhiza) promote osteogenesis of hMSCs and periodontal ligament cells, angiogenesis, inhibition of adipogenesis [123] - [133].

Epimedium pubescens has osteogenic activity [134], Ocimum basilicum increases osteonectin and osteocalcin levels [135], Glycyrrhiza glabra (licorice root) enhances proliferation and osteogenic differentiation [136].

Foeniculum vulgare extract has effect on osteogenesis in hMSCs [137], Thymbra spicata varintricata and Cissus quadrangularis (linn) both induce mesenchymal stem cell proliferation and osteogenic differentiation [138] [139]. Propolis proved its effect in improving the bone healing [140] - [145], Camellia sinensis and acemannan (aloe vera) induce osteogenic activity and cellular proliferation and differentiation, and periodontal regeneration [85] [86] [87] [88] [89] [94] [146] [147].

Curcuma longa (curcumin) enhances cell’s proliferation, viability and bone formation [148] [149] [150] [151], Coffea arabica through chlorogenic acid induces osteogenic differentiation [152] [153] [154] [155], Kanroin ganluyin reduces osteoclast differentiation in vitro and prevents alveolar bone resorption [156], Nicotiana benthamiana induces osteogenic differentiation [157] [158] and Coconut oil induces periodontal tissue regeneration via expressions of tnf-α and tgf-β1 [159].

3.5. Action on Oral Mucosa (Wound Healing)

The wound healing process is distinguished by four phases, which are respectively, hemostasis, inflammation, proliferation, and remodeling. The physiology of wound healing is a complex biological and molecular process of recovering the normal structure and functions of injured tissues. This process includes several phases, such as inflammation which consists of hemostasis and inflammation of the tissue, then the proliferation, where the angiogenesis and the construction of an extracellular matrix are the main steps with the intervention of several cellular mediators. Finally, the wound closed, and the healthy tissue recovered during the remodeling phase [199] [200] [201].

Findings in literature proved that medicinal plants are widely used in the treatment of wounds (Table 4).

Rosmarinus officinalis has antimicrobial action, effective against gingivitis and periodontitis, accelerate the healing process. [202] [203] [204], Moringa extract, is antitumoral, stimulation of hMSCs proliferation [204] - [212], Nigella sativa (black seed or black cumin) based on the systematic review conducted by Nordin 2019 [213] balance between wound healing and tissue fibrosis depending on the state of inflammation, several studies had proven its effects in accelerating the healing process [214] - [219]. Curcumin, hamamelis virginiana, seaweed extract, thyme oil (thymus vulgaris), thymol oil (ac: 2-isopropyl-5-methyl phenol), macrotyloma uniflorum, triphala (catechin), tecomella undulate, clover honey, centella asiatica, myrica rubra all these medicinal plants are responsible for providing bioactive components that would induce tissue regeneration in a review conducted by Das et al. (2016) [220], Camellia sinensis was evaluated for its anti-inflammatory effects [221] [222] [223] [224] [225], Ziziphus mauritiana (bidara leaf) induce gingival wound healing [226] [227]. Achillea millefolium has effects on arachidonic acid metabolism and has anti-inflammatory and healing properties [228] - [234]. Malva sylvestris promotes granulation tissue formation, increases collagen synthesis and reduces fibrosis, reducing healing time and has anti-inflammatory effects in burn wounds [229] [232] [234] [235], a clinical trial was conducted to establish its pharmacological potential in the reduction of the plaque control record and gingival index [236].

Salvia officinalis and Casearia sylvestris, they both have angiogenesis properties accelerate healing process [229] [237] [238] [239], Propolis increases migration and proliferation of fibroblasts, anti-inflammatory & anti-microbial actions [63] [68] [70] [240] - [249], Aloe vera, increases type III collagen synthesis, induces the synthesis of hyaluronic acid. Induces granulation tissue formation in the remodeling phase increases wound contraction and increases macrophage

Table 4. Summary based on the literature review of the medicinal plants studied with an action on wound healing.

activation markers [217] [250] - [258], Scrophularia striata decreases the number of lymphocytes and enhanced the number of fibroblasts also the maturaty of collagen fibers and showed an additional angiogenic action [259] [260] [261] [262] [263]. Alternanthera sessilis has showed an effect on cell migration of human dermal fibroblasts and keratinocytes [264] [265] [266].

Euphorbiaceae species accelerates the rate of wound healing process and showed 100% wound contraction [267], Abelmoschus esculentus okra & Amla (emblic myrobalan) showed benefic effects on gingivitis, antibacterial effect and stabilizing collagen [268] [269].

Bilberry fruit (vaccinium myrtillus), Hawthorn berry (crateagus oxycanthus) and Liquorice root (glycyrrhiza glabral), Neem extract, Mango leaf, the Miswak (miswaak, siwak, sewak), Sesame oil, Yellow dock root, Alfalfa leaf, Cinnamon bark and Turmeric root increase the angiogenesis during the wound healing in post extraction sockets [268].

Roots of albizzia lebbeck have anti-bacterial properties which may be attributed to the enhanced collagen synthesis and a potential antioxidant activity but an evaluation for their toxicity is necessary [270].

4. Conclusion

The present review offers an in-depth analysis of the high potential of medicinal plants in promoting oral tissues regeneration and reparation although new pharmaceutical technology and pharmacological research should be performed in order to explore the potential for their active compounds, the appropriate doses and the delivery mode for the administration of the plants or their compounds.

Conflicts of Interest

The authors declare no conflicts of interest regarding the publication of this paper.

References

[1] (2020) Clinical Approaches in Endodontic Regeneration—Current and Emerging Therapeutic Perspectives. British Dental Journal, 228, 328.
https://doi.org/10.1038/s41415-020-1364-6
[2] Siddiqui, Z., Acevedo-Jake, A.M., Griffith, A., Kadincesme, N., Dabek, K., Hindi, D., et al. (2022) Cells and Material-Based Strategies for Regenerative Endodontics. Bioactive Materials, 14, 234-249.
https://doi.org/10.1016/j.bioactmat.2021.11.015
[3] Lacruz, R.S., Habelitz, S., Wright, J.T. and Paine, M.L. (2017) Dental Enamel Formation and Implications for Oral Health and Disease. Physiological Reviews, 97, 939-993.
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6151498/
https://doi.org/10.1152/physrev.00030.2016
[4] Ghannam, M.G., Alameddine, H. and Bordoni, B. (2022) Anatomy, Head and Neck, Pulp (Tooth). StatPearls Publishing, Treasure Island, FL.
[5] Baranova, J., Büchner, D., Gotz, W., Schulze, M. and Tobiasch, E. (2020) Tooth Formation: Are the Hardest Tissues of Human Body Hard to Regenerate? International Journal of Molecular Sciences, 21, Article 4031.
https://doi.org/10.3390/ijms21114031
[6] Kok, Z.Y., Alaidaroos, N.Y.A., Alraies, A., Colombo, J.S., Davies, L.C., Waddington, R.J., et al. (2022) Dental Pulp Stem Cell Heterogeneity: Finding Superior Quality “Needles” in a Dental Pulpal “Haystack” for Regenerative Medicine-Based Applications. Stem Cells International, 2022, Article ID: 9127074.
https://doi.org/10.1155/2022/9127074
[7] Waterhouse, P.J., Whitworth, J.M., Camp, J.H. and Fuks, A.B. (2011) Chapter 23. Pediatric Endodontics: Endodontic Treatment for the Primary and Young Permanent Dentition. In: Hargreaves, K.M. and Cohen, S., Eds., Cohen’s Pathways of the Pulp, 10th Edition, Mosby, St. Louis, 808-857.
https://doi.org/10.1016/B978-0-323-06489-7.00023-0
[8] Groeger, S. and Meyle, J. (2019) Oral Mucosal Epithelial Cells. Frontiers in Immunology, 10, Article 208.
https://doi.org/10.3389/fimmu.2019.00208
[9] Brizuela, M. and Winters, R. (2022) Histology, Oral Mucosa. StatPearls Publishing, Treasure Island, FL.
[10] Demarco, F., Conde, M., Cavalcanti, B., Casagrande, L., Sakai, V. and Nor, J. (2011) Dental Pulp Tissue Engineering. Brazilian Dental Journal, 22, 3-13.
https://doi.org/10.1590/S0103-64402011000100001
[11] Curson, M.E.J. and Duggal, M.S. (2003) Dental Disease | Structure of Teeth. In: Caballero, B., Ed., Encyclopedia of Food Sciences and Nutrition, 2nd Edition, Academic Press, Cambridge, MA, 1743-1746.
https://doi.org/10.1016/B0-12-227055-X/00324-2
[12] Goldberg, M., Kulkarni, A.B., Young, M. and Boskey, A. (2011) Dentin: Structure, Composition and Mineralization. Frontiers in Bioscience-Elite, 3, 711-735.
https://doi.org/10.2741/e281
[13] Wan, C., Yuan, G., Luo, D., Zhang, L., Lin, H., Liu, H., et al. (2016) The Dentin Sialoprotein (DSP) Domain Regulates Dental Mesenchymal Cell Differentiation through a Novel Surface Receptor. Scientific Reports, 6, Article No. 29666.
https://doi.org/10.1038/srep29666
[14] Yamamoto, T., Hasegawa, T., Yamamoto, T., Hongo, H. and Amizuka, N. (2016) Histology of Human Cementum: Its Structure, Function, and Development. Japanese Dental Science Review, 52, 63-74.
https://doi.org/10.1016/j.jdsr.2016.04.002
[15] Cementum—An Overview. ScienceDirect Topics.
https://www.sciencedirect.com/topics/medicine-and-dentistry/cementum
[16] Kim, M.G. and Park, C.H. (2020) Tooth-Supporting Hard Tissue Regeneration Using Biopolymeric Material Fabrication Strategies. Molecules, 25, Article 4802.
https://doi.org/10.3390/molecules25204802
[17] Nguyen, T., Mui, B., Mehrabzadeh, M., Chea, Y., Chaudhry, Z., Chaudhry, K., et al. (2013) Regeneration of Tissues of the Oral Complex: Current Clinical Trends and Research Advances. Journal of the Canadian Dental Association, 78, d1.
[18] Parham, S., Kharazi, A.Z., Bakhsheshi-Rad, H.R., Nur, H., Ismail, A.F., Sharif, S., et al. (2020) Antioxidant, Antimicrobial and Antiviral Properties of Herbal Materials. Antioxidants, 9, Article 1309.
https://doi.org/10.3390/antiox9121309
[19] Kharouf, N., Haikel, Y. and Ball, V. (2020) Polyphenols in Dental Applications. Bioengineering, 7, Article 72.
https://doi.org/10.3390/bioengineering7030072
[20] Philip, N. (2019) State of the Art Enamel Remineralization Systems: The Next Frontier in Caries Management. Caries Research, 53, 284-295.
https://doi.org/10.1159/000493031
[21] Neel, E.A.A., Aljabo, A., et al. (2016) Demineralization-Remineralization Dynamics in Teeth and Bone. International Journal of Nanomedicine, 11, 4743-4763.
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5034904/
https://doi.org/10.2147/IJN.S107624
[22] Indrapriyadharshini, K., Madan Kumar, P.D., Sharma, K. and Iyer, K. (2018) Remineralizing Potential of CPP-ACP in White Spot Lesions—A Systematic Review. Indian Journal of Dental Research, 29, 487-496.
https://doi.org/10.4103/ijdr.IJDR_364_17
[23] Cheng, L. and ten Cate, J.M. (2010) Effect of Galla chinensis on the in Vitro Remineralization of Advanced Enamel Lesions. International Journal of Oral Science, 2, 15-20.
https://doi.org/10.4248/IJOS10019
[24] Zhang, T.-T., Guo, H.-J., Liu, X.-J., Chu, J.-P. and Zhou, X.-D. (2016) Galla chinensis Compounds Remineralize Enamel Caries Lesions in a Rat Model. Caries Research, 50, 159-165.
https://doi.org/10.1159/000445036
[25] Kumar, R., Mirza, M.A., Naseef, P.P., Kuruniyan, M.S., Zakir, F. and Aggarwal, G. (2022) Exploring the Potential of Natural Product-Based Nanomedicine for Maintaining Oral Health. Molecules, 27, Article 1725.
https://doi.org/10.3390/molecules27051725
[26] Zhang, T., Chu, J. and Zhou, X. (2015) Anti-Carious Effects of Galla chinensis: A Systematic Review. Phytotherapy Research, 29, 1837-1842.
https://doi.org/10.1002/ptr.5444
[27] Sequeda-Castaneda, L., Costa, G., Celis, C., Gamboa, F., Gutierrez, S. and Luengas, P. (2016) Ilex guayusa (Aquifoliaceae): Amazon and Andean Native Plant. Pharmacologyonline, 3, 193-202.
[28] Cicciù, M., Fiorillo, L. and Cervino, G. (2019) Chitosan Use in Dentistry: A Systematic Review of Recent Clinical Studies. Marine Drugs, 17, Article 417.
https://doi.org/10.3390/md17070417
[29] Bernal, H.Y., García Martínez, H. and Quevedo Sánchez, G.F. (2011) Pautas para el conocimiento, conservación y uso sostenible de las plantas medicinales nativas en Colombia: Estrategia nacional para la conservación de plantas. Repositorio Institucional de Documentación Científica.
[30] Gutiérrez-Prieto, S.J., Sequeda-Castaneda, L.G., Penedo-Jaramillo, G.M., Chacín-Nieto, A.V., Contreras-Cáceres, D.R., Moreno-Abello, G.C., et al. (2022) In Vitro Mineral Apposition Analysis of Two Colombian Plant Extracts on Amelogenesis Imperfecta Teeth. Clinical and Experimental Dental Research, 8, 336-349.
https://doi.org/10.1002/cre2.485
[31] Reigada, J.B., Tcacenco Celize, M., Andrade, L.H., Kato, M.J., Porto, A.L.M. and Lago, J.H.G. (2007) Chemical Constituents from Piper marginatum Jacq. (Piperaceae)—Antifungal Activities and Kinetic Resolution of (RS)-Marginatumol by Candida antarctica Lipase (Novozym 435). Tetrahedron: Asymmetry, 18, 1054-1058.
https://doi.org/10.1016/j.tetasy.2007.05.006
[32] Goncalves, R., dos Santos Ayres, V.F., Magalhaes, L.G., Miller Crotti, A.E., Corrêa, G.M., Guimaraes, A.C., et al. (2019) Chemical Composition and Schistosomicidal Activity of Essential Oils of Two Piper Species from the Amazon Region. Journal of Essential Oil Bearing Plants, 22, 811-820.
https://doi.org/10.1080/0972060X.2019.1631720
[33] Brú, J. and Guzman, J.D. (2016) Folk Medicine, Phytochemistry and Pharmacological Application of Piper marginatum. Revista Brasileira de Farmacognosia, 26, 767-779.
https://doi.org/10.1016/j.bjp.2016.03.014
[34] Yamamoto, H. and Ogawa, T. (2002) Antimicrobial Activity of Perilla Seed Polyphenols against Oral Pathogenic Bacteria. Bioscience, Biotechnology, and Biochemistry, 66, 921-924.
https://doi.org/10.1271/bbb.66.921
[35] Fatimah Rahamat, S., Nor Hayati Wan Abd Manan, W., Azura Shahdan, I., Azura Jalaludin, A. and Abllah, Z. (2019) Plant-Based Milk in Arresting Caries. Materials Today: Proceedings, 16, 2231-2237.
https://doi.org/10.1016/j.matpr.2019.06.115
[36] Rahamat, S.F., Manan, W.N.H.W.A., Jalaludin, A.A. and Abllah, Z. (2019) Enamel Subsurface Remineralization Potential of Virgin Coconut Oil, Coconut Milk and Coconut Water. Materials Today: Proceedings, 16, 2238-2244.
https://doi.org/10.1016/j.matpr.2019.06.116
[37] Murray, P.E., Garcia-Godoy, F. and Hargreaves, K.M. (2007) Regenerative Endodontics: A Review of Current Status and a Call for Action. Journal of Endodontics, 33, 377-390.
https://doi.org/10.1016/j.joen.2006.09.013
[38] Xie, Z., Shen, Z., Zhan, P., Yang, J., Huang, Q., Huang, S., et al. (2021) Functional Dental Pulp Regeneration: Basic Research and Clinical Translation. International Journal of Molecular Sciences, 22, Article 8991.
https://doi.org/10.3390/ijms22168991
[39] Smojver, I., Katalinic, I., Bjelica, R., Gabric, D., Matisic, V., Molnar, V., et al. (2022) Mesenchymal Stem Cells Based Treatment in Dental Medicine: A Narrative Review. International Journal of Molecular Sciences, 23, Article 1662.
https://doi.org/10.3390/ijms23031662
[40] Kulakowski, D., Leme-Kraus, A.A., Nam, J.-W., McAlpine, J., Chen, S.-N., Pauli, G.F., et al. (2017) Oligomeric Proanthocyanidins Released from Dentin Induce Regenerative Dental Pulp Cell Response. Acta Biomaterialia, 55, 262-270.
https://doi.org/10.1016/j.actbio.2017.03.051
[41] Ding, Q., Gao, J., Zheng, J., Wang, A. and Jing, S. (2019) Astragaloside IV Attenuates Inflammatory Injury and Promotes Odontoblastic Differentiation in Lipopolysaccharide-Stimulated MDPC-23 Cells and Rat Pulpitis. Journal of Oral Pathology & Medicine, 48, 951-958.
https://doi.org/10.1111/jop.12926
[42] Prabhakar, A.R., Mandroli, P.S. and Bhat, K. (2019) Pulpotomy with Curcumin: Histological Comparison with Mineral Trioxide Aggregate in Rats. Indian Journal of Dental Research, 30, 31-36.
[43] Soudi, A., Yazdanian, M., Ranjbar, R., Tebyanian, H., Yazdanian, A., Tahmasebi, E., et al. (2021) Role and Application of Stem Cells in Dental Regeneration: A Comprehensive Overview. EXCLI Journal, 20, 454-489.
[44] Sinjari, B., Pizzicannella, J., D’Aurora, M., Zappacosta, R., Gatta, V., Fontana, A., et al. (2019) Curcumin/Liposome Nanotechnology as Delivery Platform for Anti-Inflammatory Activities via NFkB/ERK/pERK Pathway in Human Dental Pulp Treated with 2-HydroxyEthyl MethAcrylate (HEMA). Frontiers in Physiology, 10, Article 633.
https://doi.org/10.3389/fphys.2019.00633
[45] Kwon, Y.-S., Kim, H.-J., Hwang, Y.-C., Rosa, V., Yu, M.-K. and Min, K.-S. (2017) Effects of Epigallocatechin Gallate, an Antibacterial Cross-Linking Agent, on Proliferation and Differentiation of Human Dental Pulp Cells Cultured in Collagen Scaffolds. Journal of Endodontics, 43, 289-296.
https://doi.org/10.1016/j.joen.2016.10.017
[46] Mohammad, S.G., Raheel, S.A. and Baroudi, K. (2015) Histological Evaluation of Allium sativum Oil as a New Medicament for Pulp Treatment of Permanent Teeth. The Journal of Contemporary Dental Practice, 16, 85-90.
https://doi.org/10.5005/jp-journals-10024-1641
[47] Grga, D., Vesna, D. and Elena, K. (2008) The Effect of Caffeic Acid Phenethyl Ester on Healing Capacity and Repair of the Dentin-Pulp Complex: In Vivo Study. Acta Veterinaria (Beogr), 58, 99-108.
https://doi.org/10.2298/AVB0801099D
[48] Tu, M.-G., Lee, A.K.-X., Lin, Y.-H., Huang, T.-H., Ho, C.-C. and Shie, M.-Y. (2020) Caffeic Acid-Coated Nanolayer on Mineral Trioxide Aggregate Potentiates the Host Immune Responses, Angiogenesis, and Odontogenesis. Journal of Endodontics, 46, 1455-1464.
https://doi.org/10.1016/j.joen.2020.07.003
[49] Kuramoto, H., Hirao, K., Yumoto, H., Hosokawa, Y., Nakanishi, T., Takegawa, D., et al. (2019) Caffeic Acid Phenethyl Ester (CAPE) Induces VEGF Expression and Production in Rat Odontoblastic Cells. BioMed Research International, 2019, Article ID: 5390720.
https://doi.org/10.1155/2019/5390720
[50] Tand, C., Ruan, J., Zhu, Y., Li, Z., Zuo, Y. and Xu, H. (2016) [Effect of Pinus massoniana Needle Extract on Root Dentin Demineralization in Vitro]. West China Journal of Stomatology, 34, 521-525.
[51] Zhou, B., Alania, Y., Reis, M., Phansalkar, R.S., Nam, J.-W., McAlpine, J.B., et al. (2020) Tri- and Tetrameric Proanthocyanidins with Dentin Bioactivities from Pinus massoniana. The Journal of Organic Chemistry, 85, 8462-8479.
https://doi.org/10.1021/acs.joc.0c00783
[52] Aydin, B., Leme-Kraus, A.A., Vidal, C.M.P., Aguiar, T.R., Phansalkar, R.S., Nam, J.-W., et al. (2019) Evidence to the Role of Interflavan Linkages and Galloylation of Proanthocyanidins at Sustaining Long-Term Dentin Biomodification. Dental Materials, 35, 328-334.
https://doi.org/10.1016/j.dental.2018.11.029
[53] Park, C., Choi, Y.-W., Hyun, S.K., Kwon, H.J., Hwang, H.J., Kim, G.-Y., et al. (2009) Induction of G1 Arrest and Apoptosis by Schisandrin C Isolated from Schizandra chinensis Baill in Human Leukemia U937 Cells. International Journal of Molecular Medicine, 24, 495-502.
https://doi.org/10.3892/ijmm_00000258
[54] Takanche, J.S., Kim, J.-S., Kim, J.-E., Han, S.-H. and Yi, H.-K. (2018) Schisandrin C Enhances Odontoblastic Differentiation through Autophagy and Mitochondrial Biogenesis in Human Dental Pulp Cells. Archives of Oral Biology, 88, 60-66.
https://doi.org/10.1016/j.archoralbio.2018.01.018
[55] Park, S.Y., Park, S.J., Park, T.G., Rajasekar, S., Lee, S.-J. and Choi, Y.-W. (2013) Schizandrin C Exerts Anti-Neuroinflammatory Effects by Upregulating Phase II Detoxifying/Antioxidant Enzymes in Microglia. International Immunopharmacology, 17, 415-426.
https://doi.org/10.1016/j.intimp.2013.06.032
[56] Kim, S.-J., Min, H.-Y., Lee, E.J., Kim, Y.S., Bae, K., Kang, S.S., et al. (2010) Growth Inhibition and Cell Cycle Arrest in the G0/G1 by Schizandrin, a Dibenzocyclooctadiene Lignan Isolated from Schisandra chinensis, on T47D Human Breast Cancer Cells. Phytotherapy Research, 24, 193-197.
https://doi.org/10.1002/ptr.2907
[57] Vu, T.T., Nguyen, M.T., Sangvanich, P., Nguyen, Q.N. and Thunyakitpisal, P. (2020) Acemannan Used as an Implantable Biomaterial for Vital Pulp Therapy of Immature Permanent Teeth Induced Continued Root Formation. Pharmaceutics, 12, Article 644.
https://doi.org/10.3390/pharmaceutics12070644
[58] Songsiripradubboon, S., Kladkaew, S., Trairatvorakul, C., Sangvanich, P., Soontornvipart, K., Banlunara, W., et al. (2017) Stimulation of Dentin Regeneration by Using Acemannan in Teeth with Lipopolysaccharide-Induced Pulp Inflammation. Journal of Endodontics, 43, 1097-1103.
https://doi.org/10.1016/j.joen.2017.01.037
[59] Songsiripradubboon, S., Banlunara, W., Sangvanich, P., Trairatvorakul, C. and Thunyakitpisal, P. (2016) Clinical, Radiographic, and Histologic Analysis of the Effects of Acemannan Used in Direct Pulp Capping of Human Primary Teeth: Short-Term Outcomes. Odontology, 104, 329-337.
https://doi.org/10.1007/s10266-015-0215-4
[60] Sholehvar, F., Mehrabani, D., Yaghmaei, P. and Vahdati, A. (2016) The Effect of Aloe vera Gel on Viability of Dental Pulp Stem Cells. Dental Traumatology, 32, 390-396.
https://doi.org/10.1111/edt.12272
[61] Jittapiromsak, N., Sahawat, D., Banlunara, W., Sangvanich, P. and Thunyakitpisal, P. (2010) Acemannan, an Extracted Product from Aloe vera, Stimulates Dental Pulp Cell Proliferation, Differentiation, Mineralization, and Dentin Formation. Tissue Engineering Part A, 16, 1997-2006.
https://doi.org/10.1089/ten.tea.2009.0593
[62] Mohanty, S. and Ramesh, S. (2020) Comparing Quality and Quantity of Dentin Bridge Formed Using Mineral Trioxide Aggregate, Biodentine, and Propolis: A Double-Blinded Randomized Controlled Clinical Trial. World Journal of Dentistry, 11, 373-379.
https://doi.org/10.5005/jp-journals-10015-1762
[63] Shi, B., Zhao, Y. and Yuan, X. (2019) Effects of MTA and Brazilian Propolis on the Biological Properties of Dental Pulp Cells. Brazilian Oral Research, 33, e117.
https://doi.org/10.1590/1807-3107bor-2019.vol33.0117
[64] Abdel Raheem, I.A., Abdul Razek, A., Elgendy, A.A., Labah, D.A. and Saleh, N.M. (2020) Egyptian Propolis-Loaded Nanoparticles as a Root Canal Nanosealer: Sealing Ability and in Vivo Biocompatibility. International Journal of Nanomedicine, 15, 5265-5277.
https://doi.org/10.2147/IJN.S258888
[65] Kim, J.-H., Kim, S.-Y., Woo, S.-M., Jeong, H.-N., Jung, J.-Y., Kim, S.-M., et al. (2019) Combination of Mineral Trioxide Aggregate and Propolis Promotes Odontoblastic Differentiation of Human Dental Pulp Stem Cells through ERK Signaling Pathway. Food Science and Biotechnology, 28, 1801-1809.
https://doi.org/10.1007/s10068-019-00609-5
[66] El-Tayeb, M.M., Abu-Seida, A.M., El Ashry, S.H. and El-Hady, S.A. (2019) Evaluation of Antibacterial Activity of Propolis on Regenerative Potential of Necrotic Immature Permanent Teeth in Dogs. BMC Oral Health, 19, Article No. 174.
https://doi.org/10.1186/s12903-019-0835-0
[67] Ahangari, Z., Naseri, M. and Vatandoost, F. (2018) Propolis: Chemical Composition and Its Applications in Endodontics. Iranian Endodontic Journal, 13, 285-292.
[68] Abbasi, A.J., Mohammadi, F., Bayat, M., Gema, S.M., Ghadirian, H., Seifi, H., et al. (2018) Applications of Propolis in Dentistry: A Review. Ethiopian Journal of Health Sciences, 28, 505-512.
https://doi.org/10.4314/ejhs.v28i4.16
[69] Sabir, A., Mooduto, L., Kaelan, C. and Horax, S. (2017) Impact of the Use of Ethanolic Extract of Propolis, Flavonoid and Non-Flavonoid Propolis for Direct Pulp Capping in Collagen Type I Density. Brazilian Journal of Oral Sciences, 15, 264-268.
https://doi.org/10.20396/bjos.v15i4.8650037
[70] Moradi, S., Saghravanian, N., Moushekhian, S., Fatemi, S. and Forghani, M. (2015) Immunohistochemical Evaluation of Fibronectin and Tenascin Following Direct Pulp Capping with Mineral Trioxide Aggregate, Platelet-Rich Plasma and Propolis in Dogs’ Teeth. Iranian Endodontic Journal, 10, 188-192.
[71] Ahangari, Z., Naseri, M., Jalili, M., Mansouri, Y., Mashhadiabbas, F. and Torkaman, A. (2012) Effect of Propolis on Dentin Regeneration and the Potential Role of Dental Pulp Stem Cell in Guinea Pigs. Cell Journal, 13, 223-228.
[72] Sabir, A., Tabbu, C.R., Agustiono, P. and Sosroseno, W. (2005) Histological Analysis of Rat Dental Pulp Tissue Capped with Propolis. Journal of Oral Science, 47, 135-138.
https://doi.org/10.2334/josnusd.47.135
[73] Al-Shaher, A., Wallace, J., Agarwal, S., Bretz, W. and Baugh, D. (2004) Effect of Propolis on Human Fibroblasts from the Pulp and Periodontal Ligament. Journal of Endodontics, 30, 359-361.
https://doi.org/10.1097/00004770-200405000-00012
[74] Bretz, W.A., Chiego, D.J., Marcucci, M.C., Cunha, I., Custódio, A. and Schneider, L.G. (1998) Preliminary Report on the Effects of Propolis on Wound Healing in the Dental Pulp. Zeitschrift für Naturforschung, 53, 1045-1048.
https://doi.org/10.1515/znc-1998-11-1217
[75] Ilewicz, L., Szczurek, Z., Szenowski, H., Luciak, M., Ciesielski, A. and Scheller, S. (1986) [Healing of Injured Dental Pulp Covered by an Ethanol Extract of Propolis (EEP) in Light of Morphological Studies]. Czasopismo Stomatologiczne, 39, 632-642.
[76] Ilewicz, L., Scheller, S., Chruscuel, H., Bily-Pryga, Z., Korycińska-Wrońska, W. and Kalebka, P. (1982) [Further Trials of Using Ethanol-Extracted Propolis (EEP) in the Treatment of Various Diseases of the Teeth and Oral Mucosa]. Czasopismo Stomatologiczne, 35, 749-753.
[77] Martin-Gonzalez, J., Segura-Egea, J.J., Pérez-Pérez, A., Cabanillas-Balsera, D. and Sánchez-Margalet, V. (2022) Leptin in Dental Pulp and Periapical Tissues: A Narrative Review. International Journal of Molecular Sciences, 23, Article 1984.
https://doi.org/10.3390/ijms23041984
[78] Choi, S.-H., Jang, J.-H., Koh, J.-T., Chang, H.-S., Hwang, Y.-C., Hwang, I.-N., et al. (2019) Effect of Leptin on Odontoblastic Differentiation and Angiogenesis: An In Vivo Study. Journal of Endodontics, 45, 1332-1341.
https://doi.org/10.1016/j.joen.2019.08.003
[79] Lin, C.-P., Wang, Y.-L., Shen, L.-J. and Lin, C.-P. (2019) The Dentin Permeability of Anti-Inflammatory and Antibacterial Drugs: In Vitro Study. Journal of the Formosan Medical Association, 118, 828-832.
https://doi.org/10.1016/j.jfma.2018.09.009
[80] Shieh, T.-M., Hsu, S.-M., Chang, K.-C., Chen, W.-C. and Lin, D.-J. (2017) Calcium Phosphate Cement with Antimicrobial Properties and Radiopacity as an Endodontic Material. Materials, 10, Article 1256.
https://doi.org/10.3390/ma10111256
[81] Huang, M.-H., Shen, Y.-F., Hsu, T.-T., Huang, T.-H. and Shie, M.-Y. (2016) Physical Characteristics, Antimicrobial and Odontogenesis Potentials of Calcium Silicate Cement Containing Hinokitiol. Materials Science & Engineering: C, 65, 1-8.
https://doi.org/10.1016/j.msec.2016.04.016
[82] Xue, W., Yu, J. and Chen, W. (2018) Plants and Their Bioactive Constituents in Mesenchymal Stem Cell-Based Periodontal Regeneration: A Novel Prospective. BioMed Research International, 2018, Article ID: 7571363.
https://doi.org/10.1155/2018/7571363
[83] Tafazoli Moghadam, E., Yazdanian, M., Alam, M., Tebyanian, H., Tafazoli, A., Tahmasebi, E., et al. (2021) Current Natural Bioactive Materials in Bone and Tooth Regeneration in Dentistry: A Comprehensive Overview. Journal of Materials Research and Technology, 13, 2078-2114.
https://doi.org/10.1016/j.jmrt.2021.05.089
[84] Wang, Q., Wang, X. and Xu, X. (2012) Icariin: Can an Herbal Extract Enhance Dental Implant Outcomes? Dental Hypotheses, 3, 133-137.
https://doi.org/10.4103/2155-8213.106836
[85] Chantarawaratit, P., Sangvanich, P., Banlunara, W., Soontornvipart, K. and Thunyakitpisal, P. (2014) Acemannan Sponges Stimulate Alveolar Bone, Cementum and Periodontal Ligament Regeneration in a Canine Class II Furcation Defect Model. Journal of Periodontal Research, 49, 164-178.
https://doi.org/10.1111/jre.12090
[86] Le Van, C., Thi Thu, H.P., Sangvanich, P., Chuenchompoonut, V. and Thunyakitpisal, P. (2020) Acemannan Induces Rapid Early Osseous Defect Healing after Apical Surgery: A 12-Month Follow-Up of a Randomized Controlled Trial. Journal of Dental Sciences, 15, 302-309.
https://doi.org/10.1016/j.jds.2019.09.012
[87] Godoy, D.J.D., Chokboribal, J., Pauwels, R., Banlunara, W., Sangvanich, P., Jaroenporn, S., et al. (2018) Acemannan Increased Bone Surface, Bone Volume, and Bone Density in a Calvarial Defect Model in Skeletally-Mature Rats. Journal of Dental Sciences, 13, 334-341.
https://doi.org/10.1016/j.jds.2018.06.004
[88] Jansisyanont, P., Tiyapongprapan, S., Chuenchompoonut, V., Sangvanich, P., Thunyakitpisal, P. (2016) The Effect of Acemannan Sponges in Post-Extraction Socket Healing: A Randomized Trial. Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology, 28, 105-110.
https://doi.org/10.1016/j.ajoms.2015.07.006
[89] Escobedo-Lozano, A.Y., et al. (2015) Physical Properties and Antibacterial Activity of Chitosan/Acemannan Mixed Systems. Carbohydrate Polymers, 115, 707-714.
https://doi.org/10.1016/j.carbpol.2014.07.064
[90] Kunimatsu, R., Kimura, A., Sakata, S., Tsuka, Y., Yoshimi, Y., Abe, T., et al. (2022) Effects of Baicalin on the Proliferation and Expression of OPG and RANKL in Human Cementoblast-Lineage Cells. Journal of Dental Sciences, 17, 162-169.
https://doi.org/10.1016/j.jds.2021.05.009
[91] Mekhemar, M., Geib, M., Kumar, M., Radha, Hassan, Y. and Dorfer, C. (2021) Salvadora persica: Nature’s Gift for Periodontal Health. Antioxidants, 10, Article 712.
https://doi.org/10.3390/antiox10050712
[92] Zhao, B., Zhang, W., Xiong, Y., Zhang, Y., Zhang, D. and Xu, X. (2020) Effects of Rutin on the Oxidative Stress, Proliferation and Osteogenic Differentiation of Periodontal Ligament Stem Cells in LPS-Induced Inflammatory Environment and the Underlying Mechanism. Journal of Molecular Histology, 51, 161-171.
https://doi.org/10.1007/s10735-020-09866-9
[93] Reynolds, M.A., Aichelmann-Reidy, M.E. and Branch-Mays, G.L. (2010) Regeneration of Periodontal Tissue: Bone Replacement Grafts. Dental Clinics of North America, 54, 55-71.
https://doi.org/10.1016/j.cden.2009.09.003
[94] Kao, R.T., Nares, S. and Reynolds, M.A. (2015) Periodontal Regeneration—Intrabony Defects: A Systematic Review from the AAP Regeneration Workshop. Journal of Periodontology, 86, S77-S104.
https://doi.org/10.1902/jop.2015.130685
[95] Tao, K., Xiao, D., Weng, J., Xiong, A., Kang, B. and Zeng, H. (2016) Berberine Promotes Bone Marrow-Derived Mesenchymal Stem Cells Osteogenic Differentiation via Canonical Wnt/β-Catenin Signaling Pathway. Toxicology Letters, 240, 68-80.
https://doi.org/10.1016/j.toxlet.2015.10.007
[96] Xin, B.-C., Wu, Q.-S., Jin, S., Luo, A.-H., Sun, D.-G. and Wang, F. (2020) Berberine Promotes Osteogenic Differentiation of Human Dental Pulp Stem Cells through Activating EGFR-MAPK-Runx2 Pathways. PPathology and Oncology Research, 26, 1677-1685.
https://doi.org/10.1007/s12253-019-00746-6
[97] Xie, Q., Johnson, B.R., Wenckus, C.S., Fayad, M.I. and Wu, C.D. (2012) Efficacy of Berberine, an Antimicrobial Plant Alkaloid, as an Endodontic Irrigant against a Mixed-Culture Biofilm in an in Vitro Tooth Model. Journal of Endodontics, 38, 1114-1117.
https://doi.org/10.1016/j.joen.2012.04.023
[98] Dong, M., Wu, S., Xu, H., Yu, X., Wang, L., Bai, H., et al. (2021) FBS-Derived Exosomes as a Natural Nano-Scale Carrier for Icariin Promote Osteoblast Proliferation. Front Bioeng Biotechnol, 9, Article 615920.
https://doi.org/10.3389/fbioe.2021.615920
[99] Londzin, P., Kocik, S., Kisiel-Nawrot, E., Janas, A., Skoczyńska, A., Krivosíková, Z., et al. (2022) Lack of Berberine Effect on Bone Mechanical Properties in Rats with Experimentally Induced Diabetes. Biomedicine & Pharmacotherapy, 146, Article ID: 112562.
https://doi.org/10.1016/j.biopha.2021.112562
[100] Zhang, J., Zhang, D., Wu, C., Liu, A., Zhang, C., Jiao, J., et al. (2019) Icariin-Conditioned Serum Engineered with Hyaluronic Acid Promote Repair of Articular Cartilage Defects in Rabbit Knees. BMC Complementary and Alternative Medicine, 19, Article No. 155.
https://doi.org/10.1186/s12906-019-2570-0
[101] Wu, L., Xia, M., Duan, Y., Zhang, L., Jiang, H., Hu, X., et al. (2019) Berberine Promotes the Recruitment and Activation of Brown Adipose Tissue in Mice and Humans. Cell Death & Disease, 10, Article No. 468.
https://doi.org/10.1038/s41419-019-1706-y
[102] Cui, Y., Xie, J., Fu, Y., Li, C., Zheng, L., Huang, D., et al. (2020) Berberine Mediates Root Remodeling in an Immature Tooth with Apical Periodontitis by Regulating Stem Cells from Apical Papilla Differentiation. International Journal of Oral Science, 12, Article No. 18.
https://doi.org/10.1038/s41368-020-0085-7
[103] Udalamaththa, V.L., Jayasinghe, C.D. and Udagama, P.V. (2016) Potential Role of Herbal Remedies in Stem Cell Therapy: Proliferation and Differentiation of Human Mesenchymal Stromal Cells. Stem Cell Research & Therapy, 7, Article No. 110.
https://doi.org/10.1186/s13287-016-0366-4
[104] Alaribe, F.N., Razwinani, M., Maepa, M., Motaung, K.S.C., Alaribe, F.N., Razwinani, M., et al. (2019) The Potential Effect of Medicinal Plants for Cartilage Regeneration. In Nikolopoulos, D.D., Safos, G.K. and Dimitrios, K., Eds., Cartilage Tissue Engineering and Regeneration Techniques, IntechOpen, London.
[105] Abbaszadegan, A., Gholami, A., Mirhadi, H., Saliminasab, M., Kazemi, A. and Moein, M.R. (2015) Antimicrobial and Cytotoxic Activity of Ferula gummosa Plant Essential Oil Compared to NaOCl and CHX: A Preliminary in Vitro Study. Restorative Dentistry & Endodontics, 40, 50-57.
https://doi.org/10.5395/rde.2015.40.1.50
[106] Ozcan-Kucuk, A., Alan, H., Gul, M. and Yolcu, U. (2018) Evaluating the Effect of Resveratrol on the Healing of Extraction Sockets in Cyclosporine A-Treated Rats. Journal of Oral and Maxillofacial Surgery, 76, 1404-1413.
https://doi.org/10.1016/j.joms.2018.02.030
[107] Casarin, R.C., Casati, M.Z., Pimentel, S.P., Cirano, F.R., Algayer, M., Pires, P.R., et al. (2014) Resveratrol Improves Bone Repair by Modulation of Bone Morphogenetic Proteins and Osteopontin Gene Expression in Rats. International Journal of Oral and Maxillofacial Surgery, 43, 900-906.
https://doi.org/10.1016/j.ijom.2014.01.009
[108] Mmadira, M.G. and Keolebogile, M.S. (2016) The Use of Bone Morphogenetic Protein-7 and Resveratrol in Collagen Type II of Articular Cartilage. Journal of Pharmacy and Pharmacology, 4, 199-211.
[109] Moon, D.K., Kim, B.G., Lee, A.R., Choe, Y. I., Khan, I., Moon, K.M., et al. (2020) Resveratrol Can Enhance Osteogenic Differentiation and Mitochondrial Biogenesis from Human Periosteum-Derived Mesenchymal Stem Cells. Journal of Orthopaedic Surgery, 15, Article No. 203.
https://doi.org/10.1186/s13018-020-01684-9
[110] Ornstrup, M.J., Harslof, T., Kjaer, T.N., Langdahl, B.L. and Pedersen, S.B. (2014) Resveratrol Increases Bone Mineral Density and Bone Alkaline Phosphatase in Obese Men: A Randomized Placebo-Controlled Trial. The Journal of Clinical Endocrinology & Metabolism, 99, 4720-4729.
https://doi.org/10.1210/jc.2014-2799
[111] Ikeda, E., Ikeda, Y., Wang, Y., Fine, N., Sheikh, Z., Viniegra, A., et al. (2018) Resveratrol Derivative-Rich Melinjo Seed Extract Induces Healing in a Murine Model of Established Periodontitis. Journal of Periodontology, 89, 586-595.
https://doi.org/10.1002/JPER.17-0352
[112] Li, G., Zhang, X., Zhang, J., Chan, C., Yew, D.T.W., He, M., et al. (2010) Ethanol Extract of Fructus Ligustri Lucidi Promotes Osteogenesis of Mesenchymal Stem Cells. Phytotherapy Research, 24, 571-576.
https://doi.org/10.1002/ptr.2987
[113] Ko, C.H., Siu, W.S., Lau, C.P., Lau, C.S., Fung, K.P. and Leung, P.C. (2010) Osteoprotective Effects of Fructus Ligustri Lucidi Aqueous Extract in Aged Ovariectomized Rats. Chinese Medicine, 5, Article No. 39.
https://doi.org/10.1186/1749-8546-5-39
[114] Gao, L.-N., An, Y., Lei, M., Li, B., Yang, H., Lu, H., et al. (2013) The Effect of the Coumarin-Like Derivative Osthole on the Osteogenic Properties of Human Periodontal Ligament and Jaw Bone Marrow Mesenchymal Stem Cell Sheets. Biomaterials, 34, 9937-9951.
https://doi.org/10.1016/j.biomaterials.2013.09.017
[115] Zhang, Z.-R., Leung, W.N., Li, G., Kong, S.K., Lu, X., Wong, Y.M., et al. (2017) Osthole Enhances Osteogenesis in Osteoblasts by Elevating Transcription Factor Osterix via cAMP/CREB Signaling in Vitro and in Vivo. Nutrients, 9, Article No. 588.
https://doi.org/10.3390/nu9060588
[116] Sun, J., Dong, Z., Zhang, Y., He, X., Fei, D., Jin, F., et al. (2017) Osthole Improves Function of Periodontitis Periodontal Ligament Stem Cells via Epigenetic Modification in Cell Sheets Engineering. Scientific Reports, 7, Article No. 5254.
https://doi.org/10.1038/s41598-017-05762-7
[117] Wang, P., Ying, J., Luo, C., Jin, X., Zhang, S., Xu, T., et al. (2017) Osthole Promotes Bone Fracture Healing through Activation of BMP Signaling in Chondrocytes. International Journal of Biological Sciences, 13, 996-1007.
https://doi.org/10.7150/ijbs.19986
[118] Zhang, Z., Leung, W.N., Li, G., Lai, Y.M. and Chan, C.W. (2016) Osthole Promotes Endochondral Ossification and Accelerates Fracture Healing in Mice. Calcified Tissue International, 99, 649-660.
https://doi.org/10.1007/s00223-016-0189-4
[119] Zhang, J.-F., Li, G., Meng, C.-L., Dong, Q., Chan, C.-Y., He, M.-L., et al. (2009) Total Flavonoids of Herba Epimedii Improves Osteogenesis and Inhibits Osteoclastogenesis of Human Mesenchymal Stem Cells. Phytomedicine, 16, 521-529.
https://doi.org/10.1016/j.phymed.2009.01.003
[120] Zhang, P., Dai, K., Yan, S., Yan, W., Zhang, C., et al. (2009) Effects of Naringin on the Proliferation and Osteogenic Differentiation of Human Bone Mesenchymal Stem Cell. European Journal of Pharmacology, 607, 1-5.
https://doi.org/10.1016/j.ejphar.2009.01.035
[121] Sharma, A., Bhardwaj, P. and Arya, S.K. (2021) Naringin: A Potential Natural Product in the Field of Biomedical Applications. Carbohydrate Polymer Technologies and Applications, 2, Article ID: 100068.
https://doi.org/10.1016/j.carpta.2021.100068
[122] Gu, Q., Chen, C., Zhang, Z., Wu, Z., Fan, X., Zhang, Z., et al. (2015) Ginkgo biloba Extract Promotes Osteogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells in a Pathway Involving Wnt/β-Catenin Signaling. Pharmacological Research, 97, 70-78.
https://doi.org/10.1016/j.phrs.2015.04.004
[123] Chin, A., Yang, Y., Chai, L., Wong, R.W.K. and Rabie, A.-B.M. (2011) Effects of Medicinal Herb Salvia Miltiorrhiza on Osteoblastic Cells in Vitro. Journal of Orthopaedic Research, 29, 1059-1063.
https://doi.org/10.1002/jor.21376
[124] Bian, Y. and Xiang, J. (2020) Salvianolic Acid B Promotes the Osteogenic Differentiation of Human Periodontal Ligament Cells through Wnt/β-Catenin Signaling Pathway. Archives of Oral Biology, 113, Article ID: 104693.
https://doi.org/10.1016/j.archoralbio.2020.104693
[125] Wu, Y., Zhang, C., Wu, J., Han, Y. and Wu, C. (2019) Angiogenesis and Bone Regeneration by Mesenchymal Stem Cell Transplantation with Danshen in a Rabbit Model of Avascular Necrotic Femoral Head. Experimental and Therapeutic Medicine, 18, 163-171.
https://doi.org/10.3892/etm.2019.7556
[126] Ji, C., Bi, L., Li, J. and Fan, J. (2019) Salvianolic Acid B-Loaded Chitosan/hydroxyapatite Scaffolds Promotes The Repair Of Segmental Bone Defect By Angiogenesis and Osteogenesis. International Journal of Nanomedicine, 14, 8271-8284.
https://doi.org/10.2147/IJN.S219105
[127] Yang, Y., Zhu, Z., Wang, D., Zhang, X., Liu, Y., Lai, W., et al. (2018) Tanshinol Alleviates Impaired Bone Formation by Inhibiting Adipogenesis via KLF15/PPARγ2 Signaling in GIO Rats. Acta Pharmacologica Sinica, 39, 633-641.
https://doi.org/10.1038/aps.2017.134
[128] Lee, D.-H., Kim, I.-K., Cho, H.-Y., Seo, J.-H., Jang, J.-M. and Kim, J. (2018) Effect of Herbal Extracts on Bone Regeneration in a Rat Calvaria Defect Model and Screening System. Journal of the Korean Association of Oral and Maxillofacial Surgeons, 44, 79-85.
https://doi.org/10.5125/jkaoms.2018.44.2.79
[129] Han, J. and Wang, W. (2017) Effects of Tanshinol on Markers of Bone Turnover in Ovariectomized Rats and Osteoblast Cultures. PLOS ONE, 12, e0181175.
https://doi.org/10.1371/journal.pone.0181175
[130] Chen, G., Zhang, X., Lin, H., Huang, G., Chen, Y. and Cui, L. (2017) Tanshinol Alleviates Osteoporosis and Myopathy in Glucocorticoid-Treated Rats. Planta Medica, 83, 1264-1273.
https://doi.org/10.1055/s-0043-108761
[131] Yang, Y., Su, Y., Wang, D., Chen, Y., Liu, Y., Luo, S., et al. (2016) Tanshinol Rescues the Impaired Bone Formation Elicited by Glucocorticoid Involved in KLF15 Pathway. Oxidative Medicine and Cellular Longevity, 2016, Article ID: 1092746.
https://doi.org/10.1155/2016/1092746
[132] Xu, D., Xu, L., Zhou, C., Lee, W.Y.W., Wu, T., Cui, L., et al. (2014) Salvianolic Acid B Promotes Osteogenesis of Human Mesenchymal Stem Cells through Activating ERK Signaling Pathway. The International Journal of Biochemistry & Cell Biology, 51, 1-9.
https://doi.org/10.1016/j.biocel.2014.03.005
[133] Cui, L., Li, T., Liu, Y., Zhou, L., Li, P., Xu, B., et al. (2012) Salvianolic Acid B Prevents Bone Loss in Prednisone-Treated Rats through Stimulation of Osteogenesis and Bone Marrow Angiogenesis. PLOS ONE, 7, e34647.
https://doi.org/10.1371/journal.pone.0034647
[134] Wang, Q., Zhou, L., Guo, Y., Liu, G., Cheng, J. and Yu, H. (2013) Differentiation of Human Adipose-Derived Stem Cells into Neuron-Like Cells by Radix Angelicae Sinensis. Neural Regeneration Research, 8, 3353-3358.
[135] Mendi, A.H., Gokcinar Yagci, B., Sarac, N., Kiziloglu, M., Yilmaz, D. and Uckan, D. (2017) Effect of Ocimum basilicum on Mesenchymal Stem Cell Proliferation and Differentiation: Does the Effect Change According to Niches? International Journal of Secondary Metabolite, 4, 1-10.
https://doi.org/10.21448/ijsm.356244
[136] Azizsoltani, A., Piri, K., Behzad, S., Soleimani, M., Nekouei, M., Mahmoudi, Z., et al. (2018) Ethyl Acetate Extract of Licorice Root (Glycyrrhiza glabra) Enhances Proliferation and Osteogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells. Iranian Journal of Pharmaceutical Research, 17, 1057-1067.
[137] Mahmoudi, Z., Soleimani, M., Saidi, A., Khamisipour, G. and Azizsoltani, A. (2013) Effects of Foeniculum vulgare Ethanol Extract on Osteogenesis in Human Mecenchymal Stem Cells. Avicenna Journal of Phytomedicine, 3, 135-142.
[138] Mendi, A., Yagci, B.G., Kiziloglu, M., Sarac, N., Ugur, A., Yilmaz, D., et al. (2017) Thymbra spicata var. intricata Induces Mesenchymal Stem Cell Proliferation and Osteogenic Differentiation. Brazilian Archives of Biology and Technology, 60, e17160391.
https://doi.org/10.1590/1678-4324-2017160391
[139] Potu, B.K., Bhat, K.M.R., Rao, M.S., Nampurath, G.K., Chamallamudi, M.R., Nayak, S.R., et al. (2009) Petroleum Ether Extract of Cissus quadrangularis (Linn.) Enhances Bone Marrow Mesenchymal Stem Cell Proliferation and Facilitates Osteoblastogenesis. Clinics, 64, 993-998.
https://doi.org/10.1590/S1807-59322009001000010
[140] Al-Hariri, M., Eldin, T.G., Abu-Hozaifa, B. and Elnour, A. (2011) Glycemic Control and Anti-Osteopathic Effect of Propolis in Diabetic Rats. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy, 4, 377-384.
https://doi.org/10.2147/DMSO.S24159
[141] Meimandi-Parizi, A., Oryan, A., Sayahi, E. and Bigham-Sadegh, A. (2018) Propolis Extract a New Reinforcement Material in Improving Bone Healing: An in Vivo Study. International Journal of Surgery, 56, 94-101.
https://doi.org/10.1016/j.ijsu.2018.06.006
[142] Somsanith, N., Kim, Y.-K., Jang, Y.-S., Lee, Y.-H., Yi, H.-K., Jang, J.-H., et al. (2018) Enhancing of Osseointegration with Propolis-Loaded TiO2 Nanotubes in Rat Mandible for Dental Implants. Materials, 11, Article 61.
https://doi.org/10.3390/ma11010061
[143] Zohery, A.A., Meshri, S.M., Madi, M.I., Abd El Rehim, S.S. and Nour, Z.M. (2018) Egyptian Propolis Compared to Nanohydroxyapatite Graft in the Treatment of Class II Furcation Defects in Dogs. Journal of Periodontology, 89, 1340-1350.
https://doi.org/10.1002/JPER.17-0685
[144] Altan, B.A., Kara, I.M., Nalcaci, R., Ozan, F., Erdogan, S.M., Ozkut, M.M., et al. (2013) Systemic Propolis Stimulates New Bone Formation at the Expanded Suture: A Histomorphometric Study. The Angle Orthodontist, 83, 286-291.
https://doi.org/10.2319/032612-253.1
[145] Yanagita, M., Kojima, Y., Mori, K., Yamada, S. and Murakami, S. (2011) Osteoinductive and Anti-Inflammatory Effect of Royal Jelly on Periodontal Ligament Cells. Biomedical Research, 32, 285-291.
https://doi.org/10.2220/biomedres.32.285
[146] Soares, I.M.V., Fernandes, G.V. de O., Larissa Cordeiro, C., Leite, Y.K.P. de C., Bezerra, D. de O., et al. (2019) The Influence of Aloe vera with Mesenchymal Stem Cells from Dental Pulp on Bone Regeneration: Characterization and Treatment of Non-Critical Defects of the Tibia in Rats. Journal of Applied Oeal Scenece, 27, e20180103.
https://doi.org/10.1590/1678-7757-2018-0103
[147] Chu, C., Liu, L., Wang, Y., Yang, R., Hu, C., Rung, S., et al. (2019) Evaluation of Epigallocatechin-3-Gallate (EGCG)-Modified Scaffold Determines Macrophage Recruitment. Materials Science & Engineering: C, 100, 505-513.
https://doi.org/10.1016/j.msec.2019.03.007
[148] Jain, S., Krishna Meka, S.R. and Chatterjee, K. (2016) Curcumin Eluting Nanofibers Augment Osteogenesis toward Phytochemical Based Bone Tissue Engineering. Biomedical Materials, 11, Article ID: 055007.
https://doi.org/10.1088/1748-6041/11/5/055007
[149] Sarkar, N. and Bose, S. (2019) Liposome-Encapsulated Curcumin-Loaded 3D Printed Scaffold for Bone Tissue Engineering. ACS Applied Materials & Interfaces, 11, 17184-17192.
https://doi.org/10.1021/acsami.9b01218
[150] Bose, S., Sarkar, N. and Banerjee, D. (2018) Effects of PCL, PEG and PLGA Polymers on Curcumin Release from Calcium Phosphate Matrix for in Vitro and in Vivo Bone Regeneration. Materials Today Chemistry, 8, 110-120.
https://doi.org/10.1016/j.mtchem.2018.03.005
[151] Li, Y. and Zhang, Z.-Z. (2018) Sustained Curcumin Release from PLGA Microspheres Improves Bone Formation under Diabetic Conditions by Inhibiting the Reactive Oxygen Species Production. Drug Design, Development and Therapy, 12, 1453-1466.
https://doi.org/10.2147/DDDT.S154334
[152] Zhou, R.P., Lin, S.J., Wan, W.B., Zuo, H.L., Yao, F.F., Ruan, H.B., et al. (2016) Chlorogenic Acid Prevents Osteoporosis by Shp2/PI3K/Akt Pathway in Ovariectomized Rats. PLOS ONE, 11, e0166751.
https://doi.org/10.1371/journal.pone.0166751
[153] Palaniraj, S., Murugesan, R. and Narayan, S. (2019) Chlorogenic Acid-Loaded Calcium Phosphate Chitosan Nanogel as Biofilm Degradative Materials. The International Journal of Biochemistry & Cell Biology, 114, Article ID: 105566.
https://doi.org/10.1016/j.biocel.2019.105566
[154] Cheng, X., Li, K., Xu, S., Li, P., Yan, Y., Wang, G., et al. (2018) Applying Chlorogenic Acid in an Alginate Scaffold of Chondrocytes Can Improve the Repair of Damaged Articular Cartilage. PLOS ONE, 13, e0195326.
https://doi.org/10.1371/journal.pone.0195326
[155] Fujita, K., Otsuka, T., Yamamoto, N., Kainuma, S., Ohguchi, R., Kawabata, T., et al. (2017) (-)-Epigallocatechin Gallate but Not Chlorogenic Acid Upregulates Osteoprotegerin Synthesis through Regulation of Bone Morphogenetic Protein-4 in Osteoblasts. Experimental and Therapeutic Medicine, 14, 417-423.
https://doi.org/10.3892/etm.2017.4491
[156] Inagaki, Y., Kido, J.-I., Nishikawa, Y., Kido, R., Sakamoto, E., Bando, M., et al. (2021) Gan-Lu-Yin (Kanroin), Traditional Chinese Herbal Extracts, Reduces Osteoclast Differentiation in Vitro and Prevents Alveolar Bone Resorption in Rat Experimental Periodontitis. Journal of Clinical Medicine, 10, Article 386.
https://doi.org/10.3390/jcm10030386
[157] Rattanapisit, K., Abdulheem, S., Chaikeawkaew, D., Kubera, A., Mason, H.S., Ma, J.K.-C., et al. (2017) Recombinant Human Osteopontin Expressed in Nicotiana benthamiana Stimulates Osteogenesis Related Genes in Human Periodontal Ligament Cells. Scientific Reports, 7, Article No. 17358.
https://doi.org/10.1038/s41598-017-17666-7
[158] Chaikiawkeaw, D., Khorattanakulchai, N., Nammultriputtar, K., Rattanapisit, K., Everts, V., Kubera, A., et al. (2022) Osteopontin Induces Osteogenic Differentiation by Human Periodontal Ligament Cells via Calcium Binding Domain-ALK-1 Interaction. Journal of Periodontology, 93, e13-e23.
https://doi.org/10.1002/JPER.21-0184
[159] Thahir, H., Irawaty Djais, A., Nasir, M., Rahayu Feblina, A., Annisa, A., Etriyani, N., et al. (2022) Virgin Coconut Oil as a New Concept for Periodontal Tissue Regeneration via Expressions of TNF-α and TGF-β1. International Journal of Biomaterials, 2022, Article ID: 7562608.
https://doi.org/10.1155/2022/7562608
[160] Xu, H., Zhou, S., Qu, R., Yang, Y., Gong, X., Hong, Y., et al. (2020) Icariin Prevents Oestrogen Deficiency-Induced Alveolar Bone Loss through Promoting Osteogenesis via STAT3. Cell Proliferation, 53, e12743.
https://doi.org/10.1111/cpr.12743
[161] Xie, Y., Sun, W., Yan, F., Liu, H., Deng, Z. and Cai, L. (2019) Icariin-Loaded Porous Scaffolds for Bone Regeneration through the Regulation of the Coupling Process of Osteogenesis and Osteoclastic Activity. International Journal of Nanomedicine, 14, 6019-6033.
https://doi.org/10.2147/IJN.S203859
[162] Gong, M., Chi, C., Ye, J., Liao, M., Xie, W., Wu, C., et al. (2018) Icariin-Loaded Electrospun PCL/Gelatin Nanofiber Membrane as Potential Artificial Periosteum. Colloids and Surfaces B: Biointerfaces, 170, 201-209.
https://doi.org/10.1016/j.colsurfb.2018.06.012
[163] Lai, Y., Cao, H., Wang, X., Chen, S., Zhang, M., Wang, N., et al. (2018) Porous Composite Scaffold Incorporating Osteogenic Phytomolecule Icariin for Promoting Skeletal Regeneration in Challenging Osteonecrotic Bone in Rabbits. Biomaterials, 153, 1-13.
https://doi.org/10.1016/j.biomaterials.2017.10.025
[164] Li, M., Gu, Q., Chen, M., Zhang, C., Chen, S. and Zhao, J. (2017) Controlled Delivery of Icariin on Small Intestine Submucosa for Bone Tissue Engineering. Materials Science & Engineering C: Materials for Biological Applications, 71, 260-267.
https://doi.org/10.1016/j.msec.2016.10.016
[165] Yin, L., Wang, K., Lv, X., Sun, R., Yang, S., Yang, Y., et al. (2017) The Fabrication of an ICA-SF/PLCL Nanofibrous Membrane by Coaxial Electrospinning and Its Effect on Bone Regeneration in Vitro and in Vivo. Scientific Reports, 7, Article No. 8616.
https://doi.org/10.1038/s41598-017-07759-8
[166] Wu, Y., Xia, L., Zhou, Y., Xu, Y. and Jiang, X. (2015) Icariin Induces Osteogenic Differentiation of Bone Mesenchymal Stem Cells in a MAPK-Dependent Manner. Cell Proliferation, 48, 375-384.
https://doi.org/10.1111/cpr.12185
[167] Wang, F., Liu, Z., Lin, S., Lu, H. and Xu, J. (2012) Icariin Enhances the Healing of Rapid Palatal Expansion Induced Root Resorption in Rats. Phytomedicine, 19, 1035-1041.
https://doi.org/10.1016/j.phymed.2012.06.001
[168] Boonyagul, S., Banlunara, W., Sangvanich, P. and Thunyakitpisal, P. (2014) Effect of Acemannan, an Extracted Polysaccharide from Aloe vera, on BMSCs Proliferation, Differentiation, Extracellular Matrix Synthesis, Mineralization, and Bone Formation in a Tooth Extraction Model. Odontology, 102, 310-317.
https://doi.org/10.1007/s10266-012-0101-2
[169] Aljarbou, F., Almobarak, A., Binrayes, A. and Alamri, H.M. (2022) Salvadora persica’s Biological Properties and Applications in Different Dental Specialties: A Narrative Review. Evidence-Based Complementary and Alternative Medicine, 2022, Article ID: 8667687.
https://doi.org/10.1155/2022/8667687
[170] Farag, M., Abdel-Mageed, W.M., El Gamal, A.A. and Basudan, O.A. (2021) Salvadora persica L.: Toothbrush Tree with Health Benefits and Industrial Applications—An Updated Evidence-Based Review. Saudi Pharmaceutical Journal, 29, 751-763.
https://doi.org/10.1016/j.jsps.2021.05.007
[171] Malik, A., Aftab, M., Shaukat, M.S., Khalid, B., Hameed, M. and Ahmed, R.A. (2021) Comparative Clinical Effects of Salvadora persica Oral Rinse and a Phenolic Commercial Mouth Wash on Human Oral Health: An in Vivo Randomized Trial. Journal of the Pakistan Dental Association, 30, 87-93.
https://doi.org/10.25301/JPDA.302.87
[172] Al Bayaty, F.H., Zaidi, W.I.W., Abdullah, M.N.S.Z., Emad, O. and Al-Obaidi, M.M.J. (2018) Effect of Salvadora persica (Miswak) on Alveolar Bone Healing after Tooth Extraction in Rat. Journal of International Dental and Medical Research, 11, 770-777.
[173] Akhtar, J., Siddique, K.M., Bi, S. and Mujeeb, M. (2011) A Review on Phytochemical and Pharmacological Investigations of Miswak (Salvadora persica Linn). Journal of Pharmacy and Bioallied Sciences, 3, 113-117.
https://doi.org/10.4103/0975-7406.76488
[174] Zhao, B., Zhang, W., Xiong, Y., Zhang, Y., Jia, L. and Xu, X. (2020) Rutin Protects Human Periodontal Ligament Stem Cells from TNF-α Induced Damage to Osteogenic Differentiation through Suppressing mTOR Signaling Pathway in Inflammatory Environment. Archives of Oral Biology, 109, Article ID: 104584.
https://doi.org/10.1016/j.archoralbio.2019.104584
[175] Zhao, B., Xiong, Y., Zhang, Y., Jia, L., Zhang, W. and Xu, X. (2020) Rutin Promotes Osteogenic Differentiation of Periodontal Ligament Stem Cells through the GPR30-Mediated PI3K/AKT/mTOR Signaling Pathway. Experimental Biology and Medicine, 245, 552-561.
https://doi.org/10.1177/1535370220903463
[176] Zhao, B., Zhang, Y., Xiong, Y. and Xu, X. (2019) Rutin Promotes the Formation and Osteogenic Differentiation of Human Periodontal Ligament Stem Cell Sheets in Vitro. International Journal of Molecular Medicine, 44, 2289-2297.
https://doi.org/10.3892/ijmm.2019.4384
[177] Wu, A., Bao, Y., Yu, H., Zhou, Y. and Lu, Q. (2019) Berberine Accelerates Odontoblast Differentiation by Wnt/β-Catenin Activation. Cell Reprogramming, 21, 108-114.
https://doi.org/10.1089/cell.2018.0060
[178] Wang, C.-C., Wang, C.-H., Chen, H.-C., Cherng, J.-H., Chang, S.-J., Wang, Y.-W., et al. (2018) Combination of Resveratrol-Containing Collagen with Adipose Stem Cells For craniofacial Tissue-Engineering Applications. International Wound Journal, 15, 660-672.
https://doi.org/10.1111/iwj.12910
[179] Zhai, J.-L., Weng, X.-S., Wu, Z.-H. and Guo, S.-G. (2016) Effect of Resveratrol on Preventing Steroid-Induced Osteonecrosis in a Rabbit Model. Chinese Medical Journal (Engl), 129, 824-830.
https://doi.org/10.4103/0366-6999.178952
[180] Rutledge, K.E., Cheng, Q. and Jabbarzadeh, E. (2016) Modulation of Inflammatory Response and Induction of Bone Formation Based on Combinatorial Effects of Resveratrol. Journal of Nanomedicine & Nanotechnology, 7, Article ID: 1000350.
https://doi.org/10.4172/2157-7439.1000350
[181] Ornstrup, M.J., Harslof, T., Sorensen, L., Stenkjaer, L., Langdahl, B.L. and Pedersen, S.B. (2016) Resveratrol Increases Osteoblast Differentiation in Vitro Independently of Inflammation. Calcified Tissue International, 99, 155-163.
https://doi.org/10.1007/s00223-016-0130-x
[182] Wang, W., Sun, L., Zhang, P., Song, J. and Liu, W. (2014) An Anti-Inflammatory Cell-Free Collagen/Resveratrol Scaffold for Repairing Osteochondral Defects in Rabbits. Acta Biomaterialia, 10, 4983-4995.
https://doi.org/10.1016/j.actbio.2014.08.022
[183] Kamath, M.S., Ahmed, S.S.S.J., Dhanasekaran, M. and Santosh, S.W. (2014) Polycaprolactone Scaffold Engineered for Sustained Release of Resveratrol: Therapeutic Enhancement in Bone Tissue Engineering. International Journal of Nanomedicine, 9, 183-195.
https://doi.org/10.2147/IJN.S49460
[184] Lee, A.M.C., Shandala, T., Nguyen, L., Muhlhausler, B.S., Chen, K.-M., Howe, P.R., et al. (2014) Effects of Resveratrol Supplementation on Bone Growth in Young Rats and Microarchitecture and Remodeling in Ageing Rats. Nutrients, 6, 5871-5887.
https://doi.org/10.3390/nu6125871
[185] Min, S.K., Oh, J. and Park, J.-B. (2020) The Effects of Morinda citrifolia (Noni) on the Cellular Viability and Osteogenesis of Stem Cell Spheroids. Medicina (Kaunas, Lithuania), 56, Article 389.
https://doi.org/10.3390/medicina56080389
[186] Wan Osman, W.N., Che Ahmad Tantowi, N.A., Lau, S.F. and Mohamed, S. (2019) Epicatechin and Scopoletin Rich Morinda citrifolia (Noni) Leaf Extract Supplementation, Mitigated Osteoarthritis via Anti-Inflammatory, Anti-Oxidative, and Anti-Protease Pathways. Journal of Food Biochemistry, 43, e12755.
https://doi.org/10.1111/jfbc.12755
[187] Gu, H., Boonanantanasarn, K., Kang, M., Kim, I., Woo, K.M., Ryoo, H.-M., et al. (2018) Morinda citrifolia Leaf Extract Enhances Osteogenic Differentiation through Activation of Wnt/β-Catenin Signaling. Journal of Medicinal Food, 21, 57-69.
https://doi.org/10.1089/jmf.2017.3933
[188] Shalan, N.A.A.M., Mustapha, N.M. and Mohamed, S. (2017) Noni Leaf and Black Tea Enhance Bone Regeneration in Estrogen-Deficient Rats. Nutrition, 33, 42-51.
https://doi.org/10.1016/j.nut.2016.08.006
[189] Hussain, S., Tamizhselvi, R., George, L. and Manickam, V. (2016) Assessment of the Role of Noni (Morinda citrifolia) Juice for Inducing Osteoblast Differentiation in Isolated Rat Bone Marrow Derived Mesenchymal Stem Cells. International Journal of Stem Cells, 9, 221-229.
https://doi.org/10.15283/ijsc16024
[190] Zhao, D., Wang, Q., Zhao, Y., Zhang, H., Sha, N., Tang, D., et al. (2018) The Naturally Derived Small Compound Osthole Inhibits Osteoclastogenesis to Prevent Ovariectomy-Induced Bone Loss in Mice. Menopause, 25, 1459-1469.
https://doi.org/10.1097/GME.0000000000001150
[191] Honda, Y., Takeda, Y., Li, P., Huang, A., Sasayama, S., Hara, E., et al. (2018) Epigallocatechin Gallate-Modified Gelatin Sponges Treated by Vacuum Heating as a Novel Scaffold for Bone Tissue Engineering. Molecules, 23, Article 876.
https://doi.org/10.3390/molecules23040876
[192] Kawabata, T., Tokuda, H., Sakai, G., Fujita, K., Matsushima-Nishiwaki, R., et al. (2018) Repression of IGF-I-Induced Osteoblast Migration by (-)-Epigallocatechin Gallate through p44/p42 MAP Kinase Signaling. Biomedical Reports, 9, 318-326.
https://doi.org/10.3892/br.2018.1140
[193] Katsumata, Y., Kanzaki, H., Honda, Y., Tanaka, T., Yamaguchi, Y., Itohiya, K., et al. (2018) Single Local Injection of Epigallocatechin Gallate-Modified Gelatin Attenuates Bone Resorption and Orthodontic Tooth Movement in Mice. Polymers, 10, Article 1384.
https://doi.org/10.3390/polym10121384
[194] Kuroyanagi, G., Tokuda, H., Yamamoto, N., Kainuma, S., Fujita, K., Ohguchi, R., et al. (2017) (-)-Epigallocatechin Gallate Synergistically Potentiates Prostaglandin E2-Stimulated Osteoprotegerin Synthesis in Osteoblasts. Prostaglandins & Other Lipid Mediators, 128-129, 27-33.
https://doi.org/10.1016/j.prostaglandins.2017.02.001
[195] Hong, J.-Y., Yon, J., Lee, J.-S., Lee, I.-K., Yang, C., Kim, M.-S., et al. (2015) Effects of Epigallocatechin-3-Gallate on the Healing of Extraction Sockets with a Periapical Lesion: A Pilot Study in Dogs. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 103, 727-734.
https://doi.org/10.1002/jbm.b.33238
[196] Kaida, K., Honda, Y., Hashimoto, Y., Tanaka, M. and Baba, S. (2015) Application of Green Tea Catechin for Inducing the Osteogenic Differentiation of Human Dedifferentiated Fat Cells in Vitro. International Journal of Molecular Sciences, 16, 27988-28000.
https://doi.org/10.3390/ijms161226081
[197] Mah, Y.-J., Song, J.S., Kim, S.-O., Lee, J.-H., Jeon, M., Jung, U.-W., et al. (2014) The Effect of Epigallocatechin-3-Gallate (EGCG) on Human Alveolar Bone Cells Both in Vitro and in Vivo. Archives of Oral Biology, 59, 539-549.
https://doi.org/10.1016/j.archoralbio.2014.02.011
[198] Kamiya, M., Kawase, T., Kobayashi, M., Sekine, Y., Okuda, K., Nagata, M., et al. (2012) A Short-Term Preservation of Human Cultured Periosteal Sheets, Osteogenic Grafting Materials, Using a Commercial Preservation Solution Containing Epigallocatechin-3-Gallate (Theliokeep®) under Hypothermic Conditions. Biopreservation and Biobanking, 10, 245-252.
https://doi.org/10.1089/bio.2011.0051
[199] Shah, R., Domah, F., Shah, N. and Domah, J. (2020) Surgical Wound Healing in the Oral Cavity: A Review. Dental Update, 47, 135-143.
https://doi.org/10.12968/denu.2020.47.2.135
[200] Almadani, Y.H., Vorstenbosch, J., Davison, P.G. and Murphy, A.M. (2021) Wound Healing: A Comprehensive Review. Seminars in Plastic Surgery, 35, 141-144.
https://doi.org/10.1055/s-0041-1731791
[201] Belachew, T.F., Asrade, S., Geta, M. and Fentahun, E. (2020) In Vivo Evaluation of Wound Healing and Anti-Inflammatory Activity of 80% Methanol Crude Flower Extract of Hagenia abyssinica (Bruce) J. F. Gmel in Mice. Evidence-Based Complementary and Alternative Medicine, 2020, Article ID: 9645792.
https://doi.org/10.1155/2020/9645792
[202] Valones, M.A.A., Higino, J.S., Souza, P.R.E., Crovella, S., Caldas, A. de F. and Carvalho, A. de A.T. (2016) Dentifrice Containing Extract of Rosmarinus officinalis Linn.: An Antimicrobial Evaluation. Brazilian Dental Journal, 27, 497-501.
https://doi.org/10.1590/0103-6440201600672
[203] de Macedo, L.M., dos Santos, é.M., Militao, L., Tundisi, L.L., Ataide, J.A., Souto, E.B., et al. (2020) Rosemary (Rosmarinus officinalis L., Syn Salvia rosmarinus Spenn.) and Its Topical Applications: A Review. Plants, 9, Article 651.
https://doi.org/10.3390/plants9050651
[204] Toma, A.I., Fuller, J.M., Willett, N.J. and Goudy, S.L. (2021) Oral Wound Healing Models and Emerging Regenerative Therapies. Translational Research, 236, 17-34.
https://doi.org/10.1016/j.trsl.2021.06.003
[205] Rathi, B.S., Bodhankar, S.L. and Baheti, A.M. (2006) Evaluation of Aqueous Leaves Extract of Moringa oleifera Linn for Wound Healing in Albino Rats. Indian Journal of Experimental Biology, 44, 898-901.
[206] Al-Ghanayem, A.A., Alhussaini, M.S., Asad, M. and Joseph, B. (2022) Moringa oleifera Leaf Extract Promotes Healing of Infected Wounds in Diabetic Rats: Evidence of Antimicrobial, Antioxidant and Proliferative Properties. Pharmaceuticals, 15, Article 528.
https://doi.org/10.3390/ph15050528
[207] Mohammad Shafie, N., Raja Shahriman Shah, R.N.I., Krishnan, P., Abdul Haleem, N. and Tan, T.Y.C. (2022) Scoping Review: Evaluation of Moringa oleifera (Lam.) for Potential Wound Healing in in Vivo Studies. Molecules, 27, Article 5541.
https://doi.org/10.3390/molecules27175541
[208] Tofiq, S.A., Azeez, H. and Othman, H. (2021) Wound Healing Activities of Moringa oleifera Leaves Extract Cultivated in Kurdistan Region-Iraq. Jordan Journal of Biological Sciences, 14, 637-645.
https://doi.org/10.54319/jjbs/140403
[209] Ali, A., Garg, P., Goyal, R., Kaur, G., Li, X., Negi, P., et al. (2020) A Novel Herbal Hydrogel Formulation of Moringa oleifera for Wound Healing. Plants, 10, Article 25.
https://doi.org/10.3390/plants10010025
[210] Fayemi, O.E., Ekennia, A.C., Katata-Seru, L., Ebokaiwe, A.P., Ijomone, O.M., Onwudiwe, D.C., et al. (2018) Antimicrobial and Wound Healing Properties of Polyacrylonitrile-Moringa Extract Nanofibers. ACS Omega, 3, 4791-4797.
https://doi.org/10.1021/acsomega.7b01981
[211] Fernandes, E., Pulwale, A., Patil, G. and Moghe, A. (2016) Probing Regenerative Potential of Moringa oleifera Aqueous Extracts Using in Vitro Cellular Assays. Pharmacognosy Research, 8, 231-237.
https://doi.org/10.4103/0974-8490.188877
[212] Muhammad, A.A., Pauzi, N.A.S., Arulselvan, P., Abas, F. and Fakurazi, S. (2013) In Vitro Wound Healing Potential and Identification of Bioactive Compounds from Moringa oleifera Lam. BioMed Research International, 2013, Article ID: 974580.
https://doi.org/10.1155/2013/974580
[213] Nordin, A., Kamal, H., Yazid, M.D., Saim, A. and Idrus, R. (2019) Effect of Nigella sativa and Its Bioactive Compound on Type 2 Epithelial to Mesenchymal Transition: A Systematic Review. BMC Complementary and Alternative Medicine, 19, Article No. 290.
https://doi.org/10.1186/s12906-019-2706-2
[214] Ab Rahman, M.R., Abdul Razak, F. and Mohd Bakri, M. (2014) Evaluation of Wound Closure Activity of Nigella sativa, Melastoma malabathricum, Pluchea indica, and Piper sarmentosum Extracts on Scratched Monolayer of Human Gingival Fibroblasts. Evidence-Based Complementary and Alternative Medicine, 2014, Article ID: 190342.
https://doi.org/10.1155/2014/190342
[215] Sallehuddin, N., Nordin, A., Bt Hj Idrus, R. and Fauzi, M.B. (2020) Nigella sativa and Its Active Compound, Thymoquinone, Accelerate Wound Healing in an in Vivo Animal Model: A Comprehensive Review. International Journal of Environmental Research and Public Health, 17, Article 4160.
https://doi.org/10.3390/ijerph17114160
[216] Nourbar, E., Mirazi, N., Yari, S., Rafieian-Kopaei, M. and Nasri, H. (2019) Effect of Hydroethanolic Extract of Nigella sativa L. on Skin Wound Healing Process in Diabetic Male Rats. International Journal of Preventive Medicine, 10, 18.
https://doi.org/10.4103/ijpvm.IJPVM_276_18
[217] Sari, Y., Purnawan, I., Kurniawan, D.W. and Sutrisna, E. (2018) A Comparative Study of the Effects of Nigella sativa Oil Gel and Aloe vera Gel on Wound Healing in Diabetic Rats. Journal of Evidence-Based Integrative Medicine, 23.
https://doi.org/10.1177/2515690X18772804
[218] Javadi, S.M.R., Hashemi, M., Mohammadi, Y., MamMohammadi, A., Sharifi, A. and Makarchian, H.R. (2018) Synergistic Effect of Honey and Nigella sativa on Wound Healing in Rats. Acta Cirúrgica Brasileira, 33, 518-523.
https://doi.org/10.1590/s0102-865020180060000006
[219] Han, M., Durmus, A., Sagliyan, A., Günay, C., Ozkaraca, M., Kandemir, F., et al. (2017) Effects of Nigella sativa and Hypericum perforatum on Wound Healing. Turkish Journal of Veterinary & Animal Sciences, 41, 99-105.
https://doi.org/10.3906/vet-1603-26
[220] Das, U., Behera, S.S., Singh, S., Rizvi, S.I. and Singh, A.K. (2016) Progress in the Development and Applicability of Potential Medicinal Plant Extract-Conjugated Polymeric Constructs for Wound Healing and Tissue Regeneration: Plant Molecule Conjugated Polymeric Scaffolds for Would Healing. Phytotherapy Research, 30, 1895-1904.
https://doi.org/10.1002/ptr.5700
[221] Goenka, P., Sarawgi, A., Karun, V., Nigam, A.G., Dutta, S. and Marwah, N. (2013) Camellia sinensis (Tea): Implications and Role in Preventing Dental Decay. Pharmacognosy Reviews, 7, 152-156.
https://doi.org/10.4103/0973-7847.120515
[222] Tafazoli, A. and Tafazoli Moghadam, E. (2020) Camellia sinensis Mouthwashes in Oral Care: A Systematic Review. Journal of Denistry, 21, 249-262.
[223] Hashemipour, M.A., Lotfi, S., Torabi, M., Sharifi, F., Ansari, M., Ghassemi, A., et al. (2017) Evaluation of the Effects of Three Plant Species (Myrtus communis L., Camellia sinensis L., Zataria multiflora Boiss.) on the Healing Process of Intraoral Ulcers in Rats. Journal of Dentistry, 18, 127-135.
[224] Shahbaz, M., Zaheer, N., Sagheer, A., Arshad, A.I., Zaheer, U. and Alam, M.K. (2017) Role of Green Tea Extract (Camellia sinensis) in Prevention of Nicotine-Induced Vascular Changes in Buccal Mucosa of Albino Rats. International Medical Journal, 24, 230-233.
[225] Hajiaghaalipour, F., Kanthimathi, M.S., Abdulla, M.A. and Sanusi, J. (2013) The Effect of Camellia sinensis on Wound Healing Potential in an Animal Model. Evidence-Based Complementary and Alternative Medicine, 2013, Article ID: 386734.
https://doi.org/10.1155/2013/386734
[226] Ma’ruf, M.T., Dewi, P.S. and Nurlitasari, D.F. (2021) Efficacy of Bidara Leaf (Ziziphus mauritiana) Viscous Extract to Gingival Wound Healing in Wistar Rats. Journal of International Dental & Medical Research, 14, 1367-1372.
[227] Aziza, N., Hutami, I.R., Indraswary, R. and Suryono (2022) Antibacterial Effects of Ethanolic Extract of Bidara (Ziziphus mauritiana Lam) Leaf against Porphyromonas gingivalis. Insisiva Dental Journal, 11, 62-69.
https://doi.org/10.18196/di.v11i2.15222
[228] Benedek, B., Kopp, B. and Melzig, M.F. (2007) Achillea millefolium L. s.l.—Is the Anti-Inflammatory Activity Mediated by Protease Inhibition? Journal of Ethnopharmacology, 113, 312-317.
https://doi.org/10.1016/j.jep.2007.06.014
[229] Medellín-Luna, M.F., Castaneda-Delgado, J.E., Martínez-Balderas, V.Y. and Cervantes-Villagrana, A.R. (2019) Medicinal Plant Extracts and Their Use as Wound Closure Inducing Agents. Journal of Medicinal Food, 22, 435-443.
https://doi.org/10.1089/jmf.2018.0145
[230] Hajhashemi, M., Ghanbari, Z., Movahedi, M., Rafieian, M., Keivani, A. and Haghollahi, F. (2018) The Effect of Achillea millefolium and Hypericum perforatum Ointments on Episiotomy Wound Healing in Primiparous Women. The Journal of Maternal-Fetal & Neonatal Medicine, 31, 63-69.
https://doi.org/10.1080/14767058.2016.1275549
[231] Dorjsembe, B., Lee, H.J., Kim, M., Dulamjav, B., Jigjid, T. and Nho, C.W. (2017) Achillea asiatica Extract and Its Active Compounds Induce Cutaneous Wound Healing. Journal of Ethnopharmacology, 206, 306-314.
https://doi.org/10.1016/j.jep.2017.06.006
[232] Nasiri, E., Hosseinimehr, S.J., Azadbakht, M., Akbari, J., Enayati-fard, R. and Azizi, S. (2015) Effect of Malva sylvestris Cream on Burn Injury and Wounds in Rats. Avicenna Journal of Phytomedicine, 5, 341-354.
[233] Akkol, E.K., Koca, U., Pesin, I. and Yilmazer, D. (2011) Evaluation of the Wound Healing Potential of Achillea biebersteinii Afan. (Asteraceae) by in Vivo Excision and Incision Models. Evidence-Based Complementary and Alternative Medicine, 2011, Article ID: 474026.
https://doi.org/10.1093/ecam/nep039
[234] Pirbalouti, A.G., Koohpayeh, A. and Karimi, I. (2010) The Wound Healing Activity of Flower Extracts of Punica granatum and Achillea kellalensis in Wistar Rats. Acta Poloniae Pharmaceutica, 67, 107-110.
[235] Afshar, M., Ravarian, B., Zardast, M., Moallem, S.A., Fard, M.H. and Valavi, M. (2015) Evaluation of Cutaneous Wound Healing Activity of Malva sylvestris Aqueous Extract in BALB/c Mice. Iranian Journal of Basic Medical Sciences, 18, 616-622.
[236] Aravena, P. (2021) Clinical Evaluation of a Mouthwash Containing Malva sylvestris Extract and Its Role in Reducing Oral Biofilm and Gingival Inflammation. A Randomized, Triple Masked Clinical Trial. clinicaltrials.gov.
[237] Oberlies, N.H., Burgess, J.P., Navarro, H.A., Pinos, R.E., Fairchild, C.R., Peterson, R.W., et al. (2002) Novel Bioactive Clerodane Diterpenoids from the Leaves and Twigs of Casearia sylvestris. Journal of Natural Products, 65, 95-99.
https://doi.org/10.1021/np010459m
[238] Qnais, E.Y., Abu-Dieyeh, M., Abdulla, F.A. and Abdalla, S.S. (2010) The Antinociceptive and Anti-Inflammatory Effects of Salvia officinalis Leaf Aqueous and Butanol Extracts. Pharmaceutical Biology, 48, 1149-1156.
https://doi.org/10.3109/13880200903530763
[239] de Mattos, E.S., Frederico, M.J.S., Colle, T.D., de Pieri, D.V., Peters, R.R. and Piovezan, A.P. (2007) Evaluation of Antinociceptive Activity of Casearia sylvestris and Possible Mechanism of Action. Journal of Ethnopharmacology, 112, 1-6.
https://doi.org/10.1016/j.jep.2007.01.034
[240] Barroso, P.R., Lopes-Rocha, R., Pereira, E.M.F., Marinho, S.A., de Miranda, J.L., Lima, N.L., et al. (2012) Effect of Propolis on Mast Cells in Wound Healing. Inflammopharmacology, 20, 289-294.
https://doi.org/10.1007/s10787-011-0105-5
[241] Marizela, S., Semir, S., Azra, A.-M., Midhat, J. and Zerina, B. (2019) Impact of Propolis on the Oral Health. Balkan Journal of Dental Medicine, 23, 1-9.
https://balkandentaljournal.com/impact-of-propolis-on-the-oral-health/
https://doi.org/10.2478/bjdm-2019-0001
[242] Zulhendri, F., Felitti, R., Fearnley, J. and Ravalia, M. (2021) The Use of Propolis in Dentistry, Oral Health, and Medicine: A Review. Journal of Oral Biosciences, 63, 23-34.
https://doi.org/10.1016/j.job.2021.01.001
[243] Saeed, M.A., Khabeer, A., Faridi, M.A. and Makhdoom, G. (2021) Effectiveness of Propolis in Maintaining Oral Health: A Scoping Review. Canadian Journal of Dental Hygiene, 55, 167-176.
[244] Afkhamizadeh, M., Aboutorabi, R., Ravari, H., Fathi Najafi, M., Ataei Azimi, S., Javadian Langaroodi, A., et al. (2018) Topical Propolis Improves Wound Healing in Patients with Diabetic Foot Ulcer: A Randomized Controlled Trial. Natural Product Research, 32, 2096-2099.
https://doi.org/10.1080/14786419.2017.1363755
[245] Oryan, A., Alemzadeh, E. and Moshiri, A. (2018) Potential Role of Propolis in Wound Healing: Biological Properties and Therapeutic Activities. Biomedicine & Pharmacotherapy, 98, 469-483.
https://doi.org/10.1016/j.biopha.2017.12.069
[246] Pobiega, K., Gniewosz, M. and Krasniewska, K. (2017) Antimicrobial and Antiviral Properties of Different Types of Propolis. Zeszyty Problemowe Postepów Nauk Rolniczych, No. 589, 69-79.
https://doi.org/10.22630/ZPPNR.2017.589.22
[247] Takzaree, N., Hadjiakhondi, A., Hassanzadeh, G., Rouini, M.R. and Manayi, A. (2016) Synergistic Effect of Honey and Propolis on Cutaneous Wound Healing in Rats. Acta Medica Iranica, 54, 233-239.
[248] Jacob, A., Parolia, A., Pau, A. and Davamani Amalraj, F. (2015) The Effects of Malaysian Propolis and Brazilian Red Propolis on Connective Tissue Fibroblasts in the Wound Healing Process. BMC Complementary and Alternative Medicine, 15, Article No. 294.
https://doi.org/10.1186/s12906-015-0814-1
[249] Jain, S., Rai, R., Gupta, V. and Batra, M. (2014) Propolis in Oral Health: A Natural Remedy. World Journal of Pharmaceutical Sciences, 2, 90-94.
[250] Davis, R.H., Leitner, M.G., Russo, J.M. and Byrne, M.E. (1989) Wound Healing. Oral and Topical Activity of Aloe vera. Journal of the American Podiatric Medical Association, 79, 559-562.
https://doi.org/10.7547/87507315-79-11-559
[251] Ali, F., Wajid, N., Sarwar, M.G. and Qazi, A.M. (2021) Oral Administration of Aloe vera Ameliorates Wound Healing through Improved Angiogenesis and Chemotaxis in Sprague Dawley Rats. Current Pharmaceutical Biotechnology, 22, 1122-1128.
https://doi.org/10.2174/1389201021999201001204345
[252] Sánchez, M., González-Burgos, E., Iglesias, I. and Gómez-Serranillos, M.P. (2020) Pharmacological Update Properties of Aloe vera and Its Major Active Constituents. Molecules, 25, Article 1324.
https://doi.org/10.3390/molecules25061324
[253] Jamil, D. (2018) Effect of Aloe vera on Wound Healing. Pakistan Journal of Scientific and Industrial Research, 63, 48-61.
https://doi.org/10.52763/PJSIR.BIOL.SCI.63.1.2020.48.61
[254] Tanaka, M., Misawa, E., Yamauchi, K., Abe, F. and Ishizaki, C. (2015) Effects of Plant Sterols Derived from Aloe vera Gel on Human Dermal Fibroblasts in Vitro and on Skin Condition in Japanese Women. Clinical, Cosmetic and Investigational Dermatology, 8, 95-104.
https://doi.org/10.2147/CCID.S75441
[255] Hashemi, S.A., Madani, S.A. and Abediankenari, S. (2015) The Review on Properties of Aloe vera in Healing of Cutaneous Wounds. BioMed Research International, 2015, Article ID: 714216.
https://doi.org/10.1155/2015/714216
[256] Tabandeh, M.R., Oryan, A. and Mohammadalipour, A. (2014) Polysaccharides of Aloe vera Induce MMP-3 and TIMP-2 Gene Expression during the Skin Wound Repair of Rat. International Journal of Biological Macromolecules, 65, 424-430.
https://doi.org/10.1016/j.ijbiomac.2014.01.055
[257] Budai, M.M., Varga, A., Milesz, S., Tozsér, J. and Benko, S. (2013) Aloe vera Downregulates LPS-Induced Inflammatory Cytokine Production and Expression of NLRP3 Inflammasome in Human Macrophages. Molecular Immunology, 56, 471-479.
https://doi.org/10.1016/j.molimm.2013.05.005
[258] Babaee, N., Zabihi, E., Mohseni, S. and Moghadamnia, A.A. (2012) Evaluation of the Therapeutic Effects of Aloe vera Gel on Minor Recurrent Aphthous Stomatitis. Dental Research Journal, 9, 381-385.
[259] Tanideh, N., Haddadi, M., Rokni-Hosseini, M.H., Hosseinzadeh, M., Mehrabani, D., Sayehmiri, K., et al. (2015) The Healing Effect of Scrophularia striata on Experimental Burn Wounds Infected to Pseudomonas aeruginosa in Rat. World Journal of Plastic Surgery, 4, 16-22.
[260] Chatzopoulos, G.S., Karakostas, P., Kavakloglou, S., Assimopoulou, A., Barmpalexis, P. and Tsalikis, L. (2022) Clinical Effectiveness of Herbal Oral Care Products in Periodontitis Patients: A Systematic Review. International Journal of Environmental Research and Public Health, 19, Article 10061.
https://doi.org/10.3390/ijerph191610061
[261] Kerdar, T., Rabienejad, N., Alikhani, Y., Moradkhani, S. and Dastan, D. (2019) Clinical, in Vitro and Phytochemical, Studies of Scrophularia striata Mouthwash on Chronic Periodontitis Disease. Journal of Ethnopharmacology, 239, Article ID: 111872.
https://doi.org/10.1016/j.jep.2019.111872
[262] Haddadi, R., Tamri, P. and Javani Jooni, F. (2019) In Vitro Wound Healing Activity of Scrophularia striata Hydroalcoholic Extract. South African Journal of Botany, 121, 505-509.
https://doi.org/10.1016/j.sajb.2019.01.002
[263] Ghashghaii, A., Hashemnia, M., Nikousefat, Z., Zangeneh, M.M. and Zangeneh, A. (2017) Wound Healing Potential of Methanolic Extract of Scrophularia striata in Rats. Pharmaceutical Sciences, 23, 256-263.
https://doi.org/10.15171/PS.2017.38
[264] Jalalpure, S., Agrawal, N., Patil, M., Chimkode, R. and Tripathi, A. (2008) Antimicrobial and Wound Healing Activities of Leaves of Alternanthera sessilis Linn. International Journal of Green Pharmacy, 2, 141-144.
[265] Muniandy, K., Gothai, S., Tan, W.S., Kumar, S.S., Mohd Esa, N., Chandramohan, G., et al. (2018) In Vitro Wound Healing Potential of Stem Extract of Alternanthera sessilis. Evidence-Based Complementary and Alternative Medicine, 2018, Article ID: 3142073.
https://doi.org/10.1155/2018/3142073
[266] Enechi, O.C., Odo, C.E. and Wuave, C.P. (2013) Evaluation of the in Vitro Anti-Oxidant Activity of Alternanthera brasiliana Leaves. Journal of Pharmacy Research, 6, 919-924.
https://doi.org/10.1016/j.jopr.2013.09.006
[267] Ahmed, S., Yousaf, M., Mothana, R.A. and Al-Rehaily, A.J. (2016) Studies on Wound Healing Activity of Some Euphorbia Species on Experimental Rats. African Journal of Traditional, Complementary and Alternative Medicines, 13, 145-152.
https://doi.org/10.21010/ajtcam.v13i5.19
[268] Singh, A. and Purohit, B. (2011) Tooth Brushing, Oil Pulling and Tissue Regeneration: A Review of Holistic Approaches to Oral Health. Journal of Ayurveda and Integrative Medicine, 2, 64-68.
[269] Luthfi, M., Juliastuti, W.S. and Risky, Y.A. (2020) Angiogenesis of Extracted Tooth Wound on Wistar Rats after Application of Okra (Abelmoschus esculentus) Gel Extract. Pesquisa Brasileira em Odontopediatria e Clínica Integrada, 20, 1-8.
https://doi.org/10.1590/pboci.2020.037
[270] Joshi, A., Sengar, N., Prasad, S., Goel, R., Singh, A. and Hemalatha, S. (2013) Wound-Healing Potential of the Root Extract of Albizzia lebbeck. Planta Medica, 79, 737-743.
https://doi.org/10.1055/s-0032-1328539
[271] Eyarefe, O.D., Idowu, A. and Afolabi, J.M. (2015) Healing Potentials of Oral Moringa oleifera Leaves Extract and Tetracycline on Methicillin Resistant Staphylococcus aureus Infected Wounds of Wistar Rats. Nigerian Journal of Physiological Sciences, 30, 73-78.
[272] Miyano, K., Eto, M., Hitomi, S., Matsumoto, T., Hasegawa, S., Hirano, A., et al. (2020) The Japanese Herbal Medicine Hangeshashinto Enhances Oral Keratinocyte Migration to Facilitate Healing of Chemotherapy-Induced Oral Ulcerative Mucositis. Scientific Reports, 10, Article No. 625.
https://doi.org/10.1038/s41598-019-57192-2
[273] Kim, J.-S. and Yi, H.-K. (2018) Schisandrin C Enhances Mitochondrial Biogenesis and Autophagy in C2C12 Skeletal Muscle Cells: Potential Involvement of Anti-Oxidative Mechanisms. Naunyn-Schmiedeberg’s Archives of Pharmacology, 391, 197-206.
https://doi.org/10.1007/s00210-017-1449-1

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.