Next Article in Journal
Effect of Various LED Light Qualities, Including Wide Red Spectrum-LED, on the Growth and Quality of Mini Red Romaine Lettuce (cv. Breen)
Next Article in Special Issue
Chemical Profiling and Antioxidant and Anti-Amyloid Capacities of Salvia fruticosa Extracts from Greece
Previous Article in Journal
Integrated Full-Length Transcriptome and Metabolome Profiling Reveals Flavonoid Regulation in Response to Freezing Stress in Potato
Previous Article in Special Issue
An In Vitro and In Vivo Assessment of Antitumor Activity of Extracts Derived from Three Well-Known Plant Species
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Phytochemical Compounds Involved in the Bone Regeneration Process and Their Innovative Administration: A Systematic Review

by
Alina Hanga-Farcaș
1,
Florina Miere (Groza)
2,
Gabriela Adriana Filip
3,
Simona Clichici
3,
Luminita Fritea
2,
Laura Grațiela Vicaș
4,*,
Eleonora Marian
4,
Annamaria Pallag
4,
Tunde Jurca
4,
Sanda Monica Filip
5 and
Mariana Eugenia Muresan
2
1
Doctoral School of Biomedical Science, University of Oradea, 410087 Oradea, Romania
2
Department of Preclinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 10, 1 December Square, 410073 Oradea, Romania
3
Department of Physiology, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babeș Street, 400347 Cluj-Napoca, Romania
4
Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 10, 1 December Square, 410073 Oradea, Romania
5
Department of Physics, Faculty of Informatics and Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania
*
Author to whom correspondence should be addressed.
Plants 2023, 12(10), 2055; https://doi.org/10.3390/plants12102055
Submission received: 16 April 2023 / Revised: 19 May 2023 / Accepted: 20 May 2023 / Published: 22 May 2023
(This article belongs to the Special Issue Medicinal Plant Extracts)

Abstract

:
Bone metabolism is a complex process which is influenced by the activity of bone cells (e.g., osteocytes, osteoblasts, osteoclasts); the effect of some specific biomarkers (e.g., parathyroid hormone, vitamin D, alkaline phosphatase, osteocalcin, osteopontin, osteoprotegerin, osterix, RANKL, Runx2); and the characteristic signaling pathways (e.g., RANKL/RANK, Wnt/β, Notch, BMP, SMAD). Some phytochemical compounds—such as flavonoids, tannins, polyphenols, anthocyanins, terpenoids, polysaccharides, alkaloids and others—presented a beneficial and stimulating effect in the bone regeneration process due to the pro-estrogenic activity, the antioxidant and the anti-inflammatory effect and modulation of bone signaling pathways. Lately, nanomedicine has emerged as an innovative concept for new treatments in bone-related pathologies envisaged through the incorporation of medicinal substances in nanometric systems for oral or local administration, as well as in nanostructured scaffolds with huge potential in bone tissue engineering.

1. Introduction

Bone is a complex tissue which has multiple functions and which presents a unique structure that is in permanent renewal. Bone regeneration maintains the integrity of the tissue and is coordinated by two processes: bone resorption carried out by osteoclasts, through which the removal of aged cells takes place, and the formation of new bone tissue by osteoblasts. The two processes are balanced [1]. The bone remodeling process is a continuous and cyclic one that takes place at the level of a temporary anatomical structure called the Basic Multicellular Unit (BMU) [2]. Bone remodeling at the BMU level takes place in three phases: (i) The Initiation Phase involves the selection of osteoclastic precursors and their differentiation into mature osteoclast cells and their activation for bone resorption; (ii) The Inversion Phase involves the inhibition of osteoclast activity and their apoptosis, as well as the differentiation of osteoblasts; (iii) The Terminal Phase involves the formation of new bone tissue by osteoblasts [3].
Bone formation or osteogenesis performed by the osteoblast and bone resorption or osteolysis achieved by the osteoclast are the main processes that maintain bone homeostasis. The balance between these two processes is regulated by hormonal and signaling pathways.
Parathyroid hormone (PTH) is one of the most important hormonal bone metabolism regulators. The normal activity of PTH ensures the reabsorption of calcium at the bone level. PTH binds to its receptor, type 1PTH receptor (PTHR1), which leads to the activation of protein kinase A (PKA) or protein kinase C (PKC). Using these signaling pathways, PTH can determine both bone formation and bone resorption [4]. PTHR1 is found on the surface of several cells: osteoblasts, osteoclasts and renal tubule cells; its effect on osteoblasts is a decrease in cell apoptosis, and on osteoclasts a decrease in sclerostin production [5,6]. The blood levels of PTH are directly influenced by the Ca2+ and vitamin D concentrations. Low levels of vitamin D cause hyperparathyroidism [7]. Low levels of vitamin D lead to rickets in children and osteomalacia in adults. Moreover, the use of vitamin D supplements has been correlated with an increase in calcium levels and bone density and a reduction in bone resorption and the risk of bone fractures [8,9].
In order to better understand bone metabolism, we will review the bone cells’ biomarkers involved in bone metabolism and bone regeneration and their main regulatory pathways. Additionally, the biomaterials used in bone regeneration; the plant extracts and phytochemical compounds demonstrating a positive effect on the bone regeneration process; and the pathogenetic mechanisms involved will all be described.

1.1. Bone Metabolism

1.1.1. The Main Cells of Bone Metabolism

Three main cell types are involved in the bone tissue constitution: the osteoblast, responsible for new bone formation; the osteoclast, responsible for osteolysis and osteocytes; and the mature osteoblasts, located in the bone matrix and responsible for coordinating bone hemostasis.
Osteocytes: Osteocytes are cells located at the level of the bone matrix and which are derived from mature osteoblasts [10]. They represent approximately 90–95% of bone cells, have a long life of up to 25 years, are made up of the cell body and dendrites and are found at the level of the lacuno-canalicular system, which allows their connection to adjacent osteocytes, osteoblasts, osteoclasts, bone marrow, blood vessels and nerves [11]. Osteocytes play a central role in bone homeostasis by acting as mechanical sensors and mechanical transducers for the different stimuli to which the bone is subjected. They control through chemical and hormonal responses both the process of osteolysis, by activating the cellular differentiation process of osteoclasts, and that of osteogenesis, by forming young osteocytes from osteoblasts [12,13].
Osteoblasts: Osteoblasts are small, mononucleated, cubic-shaped cells, but they can also appear in a flat or cylindrical form, and they are derived from the mesenchymal cells located in the bone marrow. Their differentiation takes place through the action of a network of cytokines and transcription factors [14]. The mesenchymal cells located at the level of the bone marrow have a multipotent character from their level and derive several cell lines: adipocytes, chondrocytes and osteoblasts [15]. Osteoblastogenesis is the cellular differentiation process of osteoblasts from mesenchymal cells. This process takes place in three phases: proliferation, matrix maturation and mineralization. The three phases are characterized by the expression of specific osteoblastic genetic markers such as: osteocalcin (OCN), osteopontin (OPN), bone sialoprotein (BSP), collagen type I (COL1A) and alkaline phosphatase (ALP). Osteoblastogenesis is controlled by hormonal factors (parathormone and glucocorticoids); vitamin D3; specific signaling pathways (Wnt, BMP, Hedgehog, Notch); circulating cytokines and transcription factors (Runx2 and Osx1); fibroblast growth factor (FGF); and transforming growth factor β (TGFβ) [16].
Osteoclasts: Osteoclasts are the cells responsible for bone resorption. They are large, multinucleated ones that are derived from hematopoietic cells, formed by the fusion of mononuclear precursors of the monocyte-macrophage line. The cellular differentiation process requires the coordination of transcription factors with co-activating and co-repressing factors [17]. In the process of cellular differentiation and maturation of osteoclasts, a central role is played by the activator receptor of nuclear factor kB ligand (RANKL). Secreted by osteoblasts, bone marrow cells and lymphocytes, RANKL is part of the tumor necrosis factor family. RANKL interacts with the RANK receptor on the cells of preosteoclasts and thus promotes their differentiation into osteoresorbing mature osteoclasts [18].

1.1.2. The Main Biomarkers of Bone Metabolism

Bone metabolism represents the sum of the biochemical processes through which osteolysis and osteogenesis take place, and it represents both the metabolism of proteins and of the minerals that enter the composition of the bone. The regulation of bone metabolism is done by a series of hormonal or enzymatic biomarkers.
Parathyroid hormone (PTH): PTH influences the biochemical processes that take place in bone remodeling. Low PTH values cause a decrease in bone circulatory markers—both osteoforming and bone resorption markers—which causes a decrease in bone turnover. By contrast, in hypoparathyroidism bone mineral density increases. The treatment of hypoparathyroidism with human parathyroid hormone (hPTH) or recombinant human parathyroid hormone (rhPTH) increases bone turnover, while bone mineral density shows increases at the hip and spine level and decreases at the radiocarpal level [19].
Vitamin D: Vitamin D deficiency causes rickets in children and osteomalacia in adults, and it is associated with the occurrence of osteoporosis and the increased risk of fractures. Recent studies have demonstrated the effect of 25-hydroxy vitamin D3 and its metabolite 1α,25 hydroxy vitamin D3 to differentiate mesenchymal stem cells of human origin towards osteoblasts [20].
Alkaline phosphatase: The bone isozyme is a glycoprotein attached to the cell membrane of osteoblasts, from where it is released into circulation, and it is the main indicator of osteogenesis. It participates in the synthesis of hydroxyapatite, providing inorganic phosphate, pyrophosphate and monophosphoesters, and at the same time it hydrolyzes pyrophosphate, which is an inhibitor of the mineralization process [21]. Tissue-nonspecific alkaline phosphatase (TNSALP) is an enzyme essential in the bone mineralization process. The mutation of the ALP gene is responsible for the synthesis of the TNSAPL enzyme and leads to hypophosphatasia [22].
Osteocalcin: Osteocalcin is the most abundant non-collagenous protein at the bone level. It is dependent on vitamin K and is produced by osteoblasts. It presents in its structure three gamma-carboxyglutamic acids that have an affinity for the Ca2+ ion, which will influence the bone remodeling and mineralization processes. It acts simultaneously as an inhibitor of the hydroxyapatite growth and as a regulator of the activity of osteoclastic precursors [23,24].
Osteopontin: Osteopontin is a phosphoprotein secreted at the bone level—especially by osteoblasts—with a role in bone metabolism, in which it participates through endocrine, neurological and immunological processes. It acts as a parathormone regulator, with low osteopontin values blocking the PTH activity of stimulating alkaline phosphatase and osteocalcin expression [25].
RANKL: RANKL is the receptor activator of NF-kB (RANK) ligand; it is a homotrimeric protein secreted by osteoblasts with functions in osteoclastogenesis [26].
Osteoprotegerin (OPG): OPG is a glycoprotein secreted specifically by osteoblasts and is a cytokine receptor of the Tumor Necrosis Factor (TNF). OPG acts as a “bait” receptor for RANKL, inhibiting osteoclastogenesis and bone resorption [27].
Osterix: Osterix is a protein with a role in the differentiation of mesenchymal cells into osteoblasts, which inhibits the formation of chondrocytes. At the osteoblastic level, osterix indicates the genetic expression of osteopotin, osteonectin, type 1a1 collagen and bone sialoprotein, which are all necessary for the process of bone mineralization at the level of osteoblasts [28].
Runx2: Runx2 is an essential protein in the maturation process of osteoblasts. Its expression is weak in mesenchymal cells and strong in immature osteoblasts. Moreover, it is essential in the differentiation of mesenchymal cells into osteoblasts [29].

1.1.3. The Main Signaling Pathways Specific to Bone Metabolism

Together with the hormonal regulation of bone homeostasis, an important role in this process belongs to the signaling pathways., as they are involved in the processes of embryonic bone development and bone repair [30].
RANKL/RANK/OPG: RANKL is produced by osteoblasts, and its binding to RANK at the level of osteoclastic precursor cells determines the differentiation of this cell line, favoring osteoclastogenesis. OPG secreted by osteoblasts is a “bait” receptor for RANKL, preventing its binding to RANK and thereby preventing osteoclastogenesis and bone resorption [26].
Wnt/β signaling: The Wnt pathway stimulates osteoblast activation and differentiation. Canonical Wnt causes β-catenin translocation and stabilization in the cell nucleus, which regulates gene transcription in response to Wnt signaling [31].
Notch: At the bone level, Notch receptors and their ligands are responsible for a multitude of phenomena: osteoblastic differentiation, bone matrix mineralization, osteoclast recruitment, cell fusion and osteoblast/osteoclast cell proliferation [32].
Bone morphogenetic proteins (BMPs): Part of the transforming growth factor β (TGFβ) family, they are involved in the processes of bone formation through the differentiation of osteoblasts. A central role in the BMP pathway signaling is played by SMAD 1, 5 and 8 proteins that interact with BMP receptors [33,34].

1.2. Biomaterials for Bone Regeneration

In many decades, a wide range of biomaterials have been developed to address bone defects promoting bone regeneration through various mechanisms, such as mechanical support, osteoconduction, osteoinduction, vascularization, neurotization, antibacterial effect, etc. [35]. Autologous bone grafts were considered the “gold standard” material for bone defects, but due to some disadvantages (limited quantity, long surgical procedures and morbidity), they have been replaced by synthetic bone grafts. Therefore, a plethora of emerging biomaterials possessing specific advantageous characteristics have been designed [36]. According to their dimensional structure and dimension size (as nanoscale), the biomaterials have been classified in zero-, one-, two-, three- and four-dimensional biomaterials [35]. Often, different classes of biomaterials have been combined, leading to hybrid composite biomaterials with synergic properties for bone tissue regeneration.
Zero-dimensional biomaterials have all three dimensions confined at nanoscale with a high surface-to-volume ratio, including some carbon-based nanomaterials (fullerene, nanodiamonds, carbon dots) and inorganic nanoparticles (NPs) (AuNPs, AgNPs, iron oxide NPs, etc.). They have presented great benefits for bone regeneration, such as biomineralization, osteogenic differentiation, good mechanical performance and biocompatibility [35].
One-dimensional biomaterials (two dimensions are nanosized) presenting high length-to-diameter ratio and unique nanotopography refer to nanowires (silicon nanowire—SiNW) and nanotubes (Titanium oxide nanotubes—TiO2NTs, carbon nanotubes—CNTs). They have modulated osteogenic and chondrogenic cell adhesion, proliferation and differentiation, and have also facilitated mineralization and demonstrated exceptional bone tissue compatibility [35].
Two-dimensional biomaterials (one dimension is in the nanoscale range), being characterized by a high diameter-to-thickness ratio, include graphene and its derivatives (graphene oxide, reduced graphene oxide). Their exceptional osteoinductive properties (in vitro and in vivo), along with their enhanced mechanical properties and favorable biocompatibility and facilitated mineralization, have been attributed to graphene due to various interactions with biomolecules and physical stress (affecting cytoskeletal tension and inducing cytoskeletal reorganization). Other nanofilm coatings (calcium phosphate coatings, black phosphorus (BP) nanofilms) have been applied to facilitate the integration of biomaterial [35].
Three-dimensional biomaterials (all dimensions are larger than the nanoscale) are the most widely used implants in clinics and include metallic (titanium and its alloy, silver, magnesium, niobium, strontium, stainless steel, cobalt, tantalum) [37]; bioceramic (bioactive bioceramics: calcium phosphate ceramics, hydroxyapatite, bioglass; bioinert bioceramics: alumina, zirconia, silicon carbide) [38]; and polymeric scaffolds and hydrogels. The disadvantages of metal-based scaffolds (poor biodegradability, local/systemic toxicity, higher elastic modulus) have been overcome by fabricating scaffolds with tunable porosity [35].
In the group of bioceramics, calcium phosphates and bioactive glass are the most frequently used in the orthopedic and dental fields. Calcium phosphates (with various forms, tunable porosities and densities) resemble the native bone tissue, having a good capacity for integration and great bioactivity and osteoconductivity [35]. Hydroxyapatite (HA) (one example of calcium phosphate) is an intrinsic component of bone tissue, presenting excellent biocompatibility, osseointegration, osteoconductivity, osteoinductivity and angiogenic effects. Biodegradable polymers have been mixed with HA in order to overcome its limitations (brittleness and insufficient mechanical strength). Bioactive glass (BG)—with its main components of silicate, borate and phosphate glass—exhibits ideal surface reactivity, bioactivity and osteoinductivity, but also brittleness (a disadvantage solved via polymer inclusion) [35].
Polymeric scaffolds are based on natural polymers (collagen, chitosan, hyaluronic acid, silk, alginate, gelatin, cellulose, etc.) and synthetic polymers (polylactic acid (PLA), poly(glycolic acid) (PGA), poly(lactic-co-glycolic acid) (PLGA), poly(ethylene glycol) (PEG), polycaprolactone (PCL), polyurethane (PU) etc.) [39,40]. Natural polymers and synthetic polymers have advantages, such as biocompatibility, design flexibility, supporting cell attachment, osteogenic differentiation, calcium biomineralization, easy tailoring of the microstructure, hydrophilicity, pore size, porosity, mechanical characteristics and degradability. They also have disadvantages, such as immunogenic and pathogenic impurities, poor replicability, need of crosslinking and an inferior loading-bearing capacity [35].
Hydrogels are based on hydrophilic polymers (natural, synthetic or hybrid) with a hydrophilic nature, with a high water content and permeability being substrates for supporting cell growth, which promotes osteogenesis, calcium biomineralization and angiogenesis [35].
Four-dimensional biomaterials are a new smart generation exhibiting a dynamic self-remodeling capability and a tunable stimuli responsiveness. They may contain hydrogels, bioceramics, piezoelectric materials, etc. that undergo self-transformation of shape and functionality after stimuli exposure [35].
The biomaterial/scaffold properties, such as pore size, stiffness, scaffold composition, surface topography, surface functional groups, surface wettability and degradation product, are crucial for their effects on cellular behavior being taken into consideration for the appropriate selection of a certain biomaterial-based scaffold [41]. The techniques applied for manufacturing the bone tissue regeneration scaffolds are freeze-drying, electrospinning, 3D printing (selective laser sintering, stereolithography, fused deposition modeling), solvent casting, sol-gel, gas foaming and particulate leaching [37]. Recently, a new 3D bioprinting technology that deposits living cells, extracellular matrices and biomaterials (inkjet-based bioprinting, extrusion-based bioprinting, laser-based bioprinting) has emerged [42].
Natural biomaterials have been widely applied in tissue regeneration due to their various advantages, such as biological and chemical similarity to natural tissues, biocompatibility, biodegradability, biological activity, low cytotoxicity, good cost-efficiency and availability. From this class, edible materials have attracted increasing interest due to some of their remarkable properties: enhancement of cell attachment, proliferation and migration and antibacterial, anti-inflammatory and antioxidant properties [43]. Edible materials, including natural polysaccharides (chitin, chitosan, hyaluronan, alginate, etc.), phenolic compounds (coumarin, phenolic acid, anthocyanin, lignin, tannic acid, etc.), and proteins (collagen, gelatin, silk fibroin, etc.) from plants, animals or other organisms, are a good source of nutrition for humans [43]. They can be employed as scaffolds or provide osteogenesis-stimulative substances for bone regeneration [43].
The inclusion of some plants, plant extracts or phytochemical compounds in the daily diet and in the biomaterials-based scaffold composition can improve the bone regeneration process. Phytochemical compounds from the classes of anthocyanins, phenols and flavonoids presented a beneficial and stimulating effect in the bone regeneration process.
Thus, the purpose of this review is to summarize the information regarding the implications of phytochemical extracts and their mechanism of action in the bone regeneration process.

2. Research Methodology

The studies considered were selected using the PRISMA 2020 flow diagram according to Page et al., 2021. The steps and selection criteria, followed by the number of the studies used for our review, are shown in Figure 1. Databases such as PubMed, Scopus, Science Direct, Elsevier, Google Scholar and Google Patents were accessed to search the literature. The Medical Subject Headings keywords included in the search were “bone regeneration”, “bone markers”, “bone signaling pathways”, “bone metabolism”, “bone pathology”, “plant extract”, “plant bone regeneration”, “natural compounds bone regeneration”, “elagic acid bone” and “bone regeneration materials”.
All the information systematized in the tables was obtained from research articles (in vivo/in vitro studies) and reviews between 2012 and 2022 (from the past 10 years). A Prisma flow-diagram was used to describe how to select the studies and articles included in the review, as shown in Figure 1 [44,45].
Studies published in languages other than English were excluded. A total of 143 studies were selected and included in this review.

3. Plant Extracts and Phytochemical Compounds with a Positive Effect on the Bone Regeneration Process

3.1. Classes of Phytochemical Compounds Involved in the Bone Regeneration Process

Natural products exhibit a wide range of modulatory effects on various pathways involved in bone regeneration (osteoclastogenesis inhibition, bone anabolism and bone resorption), such as NF-κB signaling pathways, MAPKs signaling pathways, Akt signaling pathways, calcium ion (Ca2+) signaling pathway, ROS-mediated effects and inflammatory mediator genes (Figure 2) [46,47]. Some phytochemical compounds have been effective in improving bone regeneration and preventing/treating osteoporosis and osteoarthritis by enhancing some mechanisms, such as mineral turnover, bone mineral density, inhibition of bone loss, increase in calcium and vitamin D3 and prevention of inflammation and oxidative stress [48].
The phytocompounds which have proven their potential in orthopedic applications belong to the following classes: flavonoids, tannins, polyphenols, anthocyanins, terpenoids, polysaccharides, alkaloids and others (Figure 2) [46,47,49]. Many researchers have pointed out that a certain activity is due to the complex composition of the plant product (including interactions such as the synergistic, additive, or antagonistic effect) rather than to a single compound, solvent extraction and doses [50].

3.2. Phytocompounds Used in the Bone Regeneration Process—State of the Art

Currently, many conventional treatments are known whose mechanism of action is to stop the reabsorption (anti-resorptive drugs) or anabolic drugs, but these are also known for their characteristic adverse effects following their long-term administration.
The treatment of osteoporosis can be carried out with the medicinal agents from the mentioned classes individually or as a combined treatment. A classic treatment against the loss of bone density is the combined therapy of anabolic and antiresorptive drugs, such as retiparatide, romosozumab and bisphosphonates, selective modulators of estrogen receptors, and the continuous administration of calcium combined with vitamin D. However, the demonstrated adverse effects suggest that these classical treatments should be administered with caution and not on a long-term basis. However, bone diseases such as osteoporosis and osteoarthritis are chronic diseases, so this premise is not valid.
Some of the adverse effects in the case of the long-term administration of the drug classes involved in conventional treatment are the occurrence of thrombotic events; the occurrence of breast cancer; the occurrence of kidney diseases (especially with the continuous administration of calcium together with vitamin D); gastrointestinal diseases; and cardiovascular events.
Considering these facts, the treatment plan for bone diseases—such as osteoporosis—brings to the fore natural molecules, specifically phytochemicals. These are of major interest for the treatment and stimulation of bone regeneration, and they do not cause any adverse effects.
According to the mechanism of action, natural compounds can be divided into three broad classes: compounds with pro-estrogenic activity, compounds with antioxidant and anti-inflammatory properties and modulatory compounds of bone regeneration pathways.
Estrogen has multiple effects on bone metabolism: Through its action on osteocytes, it inhibits bone remodeling; it also inhibits bone resorption through direct action on osteoclasts [51].
Oxidative stress disturbs the balance of bone metabolism, determining the apoptosis of osteocytes and osteoblasts and favoring the cell proliferation of osteoclasts. This causes bone destruction [52]. At the bone tissue level, oxidative stress favors postmenopausal, diabetic and glucocorticoid osteoporosis. Under the action of oxidative stress at the intracellular level, the mitochondria are deformed, which leads to the disruption of cellular metabolism and even to apoptosis [53].
Prostaglandin E2 (PGE2) is produced by both osteoblasts and osteoclasts in the initial phases of the bone healing process through a reaction catalyzed by the enzyme cyclooxygenase (COX). At the bone fracture, it promotes angiogenesis and increases the number of osteoclasts, which promotes osteolysis and increases the differentiation of osteoblasts, thus promoting osteogenesis. Both traditional non-steroidal anti-inflammatory drugs and selective COX2 inhibitors inhibit bone formation due to their effect on prostaglandins [54].
Proinflammatory cytokines regulate the inflammatory process of bones. They regulate both bone formation and bone resorption, thus altering bone homeostasis. Proinflammatory cytokines (TNFα, IL-1, and IL-17) cause osteoclast activation, which explains the increased bone loss during inflammation. Other cytokines, such as IL-12, IL-18, IL-33 and IFN, are suppressors of osteoclast differentiation and thus inhibit bone loss. The presence of certain cytokines in bone tissue can influence osteolysis [55].
Bone regeneration involves a large number of small molecules, transmission and signaling pathways, growth factors and physicochemical stimuli from the extracellular matrix, which are often interconnected and overlapping [56]. The main signaling pathways with a role in the bone regeneration process are Wnt, TGF-b, MAPK, JNK and the Notch pathway. Small molecules act as activators or inhibitors of transcription factors, and through this they can regulate the process of bone formation [57].
Several plant-derived components obtained from plant extracts proved their ability to affect the proliferation and differentiation potential of MSCs having as targets various signal transduction pathways demonstrating an osteopromotive role in bone regeneration [50,58,59,60]. The mechanism of action of some bioactive compounds are highlighted in Table 1.

4. The Innovative Administration and Application of Plant Extracts in the Process of Bone Regeneration

The conventional administration of treatments in bone diseases (oral, systemic administration) presents multiple disadvantages such as the low bioavailability of the administered medicinal substances or of the administered phytochemical compounds, the occurrence of side effects at the gastric and intestinal levels, low absorption and the need to increase the dose of administration [119,120,121].
Taking into account these inconveniences, traditional medicine is considered to be outdated with regards to bone diseases; therefore, the attention has been recently directed toward the targeted administration of natural compounds in nanometric form [122].
An advantage of using nanomedicine is the possibility of incorporating medicinal substances, phytochemicals or a mixture of two in different materials (biomaterials) that are compatible with bone tissue [121,122,123]. Moreover, because of their increased compatibility with the human body, the bioavailability of the substances administered through this route is increased, thus allowing a decrease in the doses administered [124,125,126].
The administration can be sustained at the level of the therapeutic plateau, and thus the number of administrations per day can be decreased. Increased compliance with the applied treatment can be thus gained [124]. The phytochemical compounds included in such systems are also protected from the factors that destroy their therapeutic activity, such as light, temperature and pH [123,124,125].
The nanometric systems also allow the controlled and targeted release of the contained compounds. The effectiveness of the treatment is much higher than that of conventional treatments [126,127,128]. Additionally, these systems allow local administration, at the bone level, which avoids the adverse gastric and intestinal effects that are encountered using conventional medication [129,130,131,132,133].
Some of these novel transport systems are presented in Figure 3.
New treatment opportunities and solutions are envisioned by combining herbal medicines having osteogenic, antitumor, antimicrobial, and anti-inflammatory properties with advanced materials for bone tissue engineering. Plant derived compounds have been included or added in the composition of orthopedic biomaterials (metallic, ceramic and polymeric matrix) in order to deliver the phytochemical substance at the bone site, leading to functionalized scaffolds with plant extracts applied in tissue engineering. A wide range of natural compounds have been proposed for incorporation within bone tissue engineering scaffolds in order to enhance bone growth, inhibit osteoclastic bone resorption, and prevent other bone-related complications [43,134,135,136]. Recently, an innovative concept has emerged in nanotechnology tissue engineering: combination and nanostructured scaffolds. These have attracted a huge interest due to their promising results in improving the bone healing process.
Essential oils (from different plant sources) and other compounds (metallic nanoparticles, zinc nitrate, copper sulphate, cobalt nitrate, etc.) have been used to treat polyurethane scaffolds, leading to the improved physical and biomedical properties of the designed scaffolds (stability, biocompatibility, bone mineralization, osteoblast cell adhesion, antimicrobial activity, etc.) [48]. Polyphenols, another class of natural compounds, have been incorporated in the composition of the bioactive scaffolds. They conferred unique structural and functional features: bio adhesion, antioxidation, anti-inflammatory and antibacterial properties, hydrophilicity, self-healing and biocompatibility (promoting bone regeneration). The approaches addressed for the fabrication of polyphenol-based scaffolds have included coating on the polymer scaffold and grafting or blending into biopolymers. They presented different morphologies, such as hydrogels (3D cross-linked networks), films (2D materials) and nanofibers (1D materials) [137].
Allium cepa extract, chitosan and poly (DL-lactic-co-glycolic) acid have been employed for the synthesis of a 3D matrix with a porous morphology (50–100 µm), allowing its surface mineralization to have a uniform hydroxyapatite layer [138]. Cucurbitacin B was incorporated into a biomaterial scaffold based on a poly (lactidecoglycolide) and β-tricalcium phosphate from where it was linearly released, showing enhanced neovascularization (via VEGFR-related signaling pathways) and bone regeneration (via higher bone mineral density, bone volume and number of trabeculae). The composite presented a bio-mimic structure with a pore between 16 and 466 nm and improved mechanical properties [114].
Aloe vera gel has been incubated on the surface of poly (3-hydroxybutyrate-co-3-hydroxyvalerate) nanofibers, generating a scaffold with promising osteoinductive potential (higher amounts of alkaline phosphatase activity, mineralization, and bone-related gene and protein expression were recorded) [139]. In another study, polycaprolactone/aloe vera/silk fibroin nanofibrous scaffolds were synthesized, followed by hydroxyapatite deposition. The result consisted of in biomimetic scaffolds with increased cell proliferation, osteogenic marker expression, osteogenic differentiation and mineralization [140].
A lipid-based self-nano emulsifying drug delivery system (100–180 nm) containing Cassia occidentalis L. butanolic extract prevented the downregulation of miR29a (Runx2), miR17 and miR20a (RANKL), induced by methyl prednisone and proving osteogenic and anti-resorptive mechanisms. The anti-inflammatory effect of the glucocorticoid was not affected, whereas the induced sarcopenia and muscle atrophy were counteracted [92]. Linum usitatissimum extract rich in phenolic compounds and flavonoids was incorporated in a composite hydrogel based on alginate and nano-hydroxyapatite which presented a porous structure (100–200 µm) and sustainedly released the natural compound. This composite demonstrated hemocompatibility, antioxidant activity and cell proliferation, thus promoting bone regeneration [141].
Fruits (grape seed, pomegranate peel, jabuticaba peel) extracts have been used as crosslinkers for anionic collagen, which together with nanohydroxyapatite formed a smart scaffold with a pore size (17–230 nm) appropriate for bone growth [142]. Genipin, a natural compound from gardenia fruits, demonstrated a promising cross-linking ability for natural biopolymers (collagen, gelatin, and chitosan) and a promising biosafety profile. The surface of several scaffolds was coated with genipin-crosslinked hydrogel releasing osteogenic factors, acting as an anti-infection agent or presenting a water-absorbing function [143].
Epigallocatechin gallate has been employed to coat poly (L-lactic acid) nanofibers to enhance hydrophilicity and stem cell adhesion, with the flavonoid serving as a protective agent of external oxidative stress for the stem cells. The plant flavonoid-based scaffold promoted the osteogenic differentiation of adipose-derived stem cells and reduced the osteoclastic maturation of the murine macrophages [101]. In another study, epigallo catechin gallate represented a promising tool to chemically modify gelatin sponges influencing surface properties (hydrophilicity and negative zeta potential). This functionalization enhanced cell adhesion and calcium phosphate precipitation, inducing superior bone formation in vivo [102]. In other study, gellan gelum hydrogels loaded with alkaline phosphatase were enriched with 5 types of gallotannins (three tannic acids with differing molecular weight, pentagalloyl glucose, and a gallotannin-rich extract from mango kernel (Mangifera indica L.). These preparations promoted the mineral formation (the dry mass percentage values were increased) due to the interactions between the ALP and gallotannins dependent on the medium [104].
Forskolin, a triterpenoid from Coleus forskohlii, was loaded into halloysite nanotubes, which were then used as a dopant for the modification biopolymer scaffold (based on gelatin, chitosan, agarose). This resulted in a new osteoconductive smart polymeric scaffold. The forskolin-loaded halloysite nanotubes acted as a filler that increased the composite volume and filled the voids, therefore facilitating the cell attachment. The mechanical properties of the scaffold together with the chemical signal of forskolin (cyclic adenosine monophosphate signaling activation in stem cells) had a synergic effect in promoting the osteodifferentiation of mesenchymal stem cells [103].
Plant-derived nanoparticles from potato containing rhamnogalacturonan-I (with relatively higher amount of gallactose) have been used for poly (L-lactide-co-ɛ-caprolactone) scaffolds functionalization revealing a downregulation of pro-inflammatory gene markers and promotion of osteogenic markers [79]. The same plant-derived pectin, rhamnogalacturonan-I, has been employed for the nanocoating of im-plants influencing osteoblast proliferation, mineralization and gene expression (Runt-related transcription factor 2 (Runx2), alkaline phosphate (ALP), osteocalcin (Bglap), α-1 type I collagen (Col1a1), receptor activator of NF-κB ligand (RANKL)) [80]. A plant-derived phenolic compound, sinapic acid, was used for the synthesis of sinapic-acid-loaded chitosan nanoparticles (100-115 nm), which were incorporated into polycaprolactone fibers (around 350 nm), resulting in a scaffold that promoted osteoblast differentiation in vitro and bone formation in vivo (via TGF-β1/BMP/Smads/Runx2 signaling pathways) due to the sustained release of sinapic acid [112].

5. Conclusions

Understanding the structure of bone tissue and the mechanisms of bone formation is crucial in the development of new methods of treatment for bone defects. The purpose of this work was to summarize the main cells, biomarkers and signaling pathways involved in bone metabolism. The phytochemical compounds with huge potential in bone regeneration were highlighted according to their activity and mechanisms of action included in three broad classes: natural compounds with pro-estrogenic activity, natural compounds with antioxidant and anti-inflammatory properties and modulatory compounds of bone signaling pathways. Recently, the targeted administration of natural compounds in nanometric forms for bone regeneration has been in high demand due to their considerable advantages. Another trend of nanomedicine applied in orthopedic field consists in the combination of plant-derived compounds with nanostructured biomaterials, resulting in functionalized scaffolds that are crucial for bone tissue engineering and that have shown promising results for the improvement of the bone healing process.

Author Contributions

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

Funding

This research was funded by University of Oradea through the grant competition “Scientific Research of Excellence Related to Priority Areas with Capitalization through Technology Transfer: INO—TRANSFER—UO”, Project no. 129/2021.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Infante, A.; Rodríguez, C.I. Osteogenesis and Aging: Lessons from Mesenchymal Stem Cells. Stem Cell Res. 2018, 9, 244. [Google Scholar] [CrossRef] [PubMed]
  2. Kim, J.H.; Kim, N. Bone Cell Communication Factors Provide a New Therapeutic Strategy for Osteoporosis. Chonnam. Med. J. 2020, 56, 94. [Google Scholar] [CrossRef]
  3. Hannah, S.S.; McFadden, S.; McNeilly, A.; McClean, C. “Take My Bone Away?” Hypoxia and Bone: A Narrative Review. J. Cell Physiol. 2021, 236, 721–740. [Google Scholar] [CrossRef] [PubMed]
  4. Wojda, S.J.; Donahue, S.W. Parathyroid Hormone for Bone Regeneration: Parathyroid Hormone For Bone Regeneration. J. Orthop. Res. 2018, 36, 2586–2594. [Google Scholar] [CrossRef]
  5. Leder, B.Z. Parathyroid Hormone and Parathyroid Hormone-Related Protein Analogs in Osteoporosis Therapy. Curr. Osteoporos. Rep. 2017, 15, 110–119. [Google Scholar] [CrossRef]
  6. Kužma, M.; Jackuliak, P.; Killinger, Z.; Payer, J. Parathyroid Hormone-Related Changes of Bone Structure. Physiol. Res. 2021, 70 (Suppl. S1), S3–S11. [Google Scholar] [CrossRef] [PubMed]
  7. Schwetz, V.; Trummer, C.; Pandis, M.; Grübler, M.; Verheyen, N.; Gaksch, M.; Zittermann, A.; März, W.; Aberer, F.; Lang, A.; et al. Effects of Vitamin D Supplementation on Bone Turnover Markers: A Randomized Controlled Trial. Nutrients 2017, 9, 432. [Google Scholar] [CrossRef] [PubMed]
  8. Lerchbaum, E.; Trummer, C.; Theiler-Schwetz, V.; Kollmann, M.; Wölfler, M.; Pilz, S.; Obermayer-Pietsch, B. Effects of Vitamin D Supplementation on Bone Turnover and Bone Mineral Density in Healthy Men: A Post-Hoc Analysis of a Randomized Controlled Trial. Nutrients 2019, 11, 731. [Google Scholar] [CrossRef]
  9. Muresan, G.C.; Hedesiu, M.; Lucaciu, O.; Boca, S.; Petrescu, N. Effect of Vitamin D on Bone Regeneration: A Review. Medicina 2022, 58, 1337. [Google Scholar] [CrossRef]
  10. Robling, A.G.; Bonewald, L.F. The Osteocyte: New Insights. Annu. Rev. Physiol. 2020, 82, 485–506. [Google Scholar] [CrossRef]
  11. Pathak, J.L.; Bravenboer, N.; Klein-Nulend, J. The Osteocyte as the New Discovery of Therapeutic Options in Rare Bone Diseases. Front. Endocrinol. 2020, 11, 405. [Google Scholar] [CrossRef]
  12. Li, M.C.M.; Chow, S.K.H.; Wong, R.M.Y.; Qin, L.; Cheung, W.H. The Role of Osteocytes-Specific Molecular Mechanism in Regulation of Mechanotransduction—A Systematic Review. J. Orthop. Transl. 2021, 29, 1–9. [Google Scholar] [CrossRef]
  13. Qin, L.; Liu, W.; Cao, H.; Xiao, G. Molecular Mechanosensors in Osteocytes. Bone Res. 2020, 8, 23. [Google Scholar] [CrossRef]
  14. Amarasekara, D.S.; Kim, S.; Rho, J. Regulation of Osteoblast Differentiation by Cytokine Networks. Int. J. Mol. Sci. 2021, 22, 2851. [Google Scholar] [CrossRef]
  15. Al-Bari, A.A.; Al Mamun, A. Current Advances in Regulation of Bone Homeostasis. FASEB BioAdvances 2020, 2, 668–679. [Google Scholar] [CrossRef] [PubMed]
  16. Ghorbaninejad, M.; Khademi-Shirvan, M.; Hosseini, S.; Baghaban Eslaminejad, M. Epidrugs: Novel Epigenetic Regulators That Open a New Window for Targeting Osteoblast Differentiation. Stem Cell Res. 2020, 11, 456. [Google Scholar] [CrossRef]
  17. Astleford, K.; Campbell, E.; Norton, A.; Mansky, K.C. Epigenetic Regulators Involved in Osteoclast Differentiation. Int. J. Mol. Sci. 2020, 21, 7080. [Google Scholar] [CrossRef]
  18. Hayashibara, T.; Hiraga, T.; Sugita, A.; Wang, L.; Hata, K.; Ooshima, T.; Yoneda, T. Regulation of Osteoclast Differentiation and Function by Phosphate: Potential Role of Osteoclasts in the Skeletal Abnormalities in Hypophosphatemic Conditions. J. Bone Min. Res. 2007, 22, 1743–1751. [Google Scholar] [CrossRef] [PubMed]
  19. Bilezikian, J.P. Hypoparathyroidism. J. Clin. Endocrinol. Metab. 2020, 105, 1722–1736. [Google Scholar] [CrossRef] [PubMed]
  20. Lou, Y.-R.; Toh, T.C.; Tee, Y.H.; Yu, H. 25-Hydroxyvitamin D3 Induces Osteogenic Differentiation of Human Mesenchymal Stem Cells. Sci. Rep 2017, 7, 42816. [Google Scholar] [CrossRef]
  21. Dempster, D.W.; Cauley, J.A.; Bouxsein, M.L.; Cosman, F. Marcus and Feldman’s Osteoporosis, 5th ed.; Dempster, D.W., Cauley, J.A., Bouxsein, M.L., Cosman, F., Eds.; Academic Press: London, UK, 2020; Volume 1, ISBN 978-0-12-813074-2. [Google Scholar]
  22. Millán, J.L.; Whyte, M.P. Alkaline Phosphatase and Hypophosphatasia. Calcif Tissue Int. 2016, 98, 398–416. [Google Scholar] [CrossRef]
  23. Moriishi, T.; Ozasa, R.; Ishimoto, T.; Nakano, T.; Hasegawa, T.; Miyazaki, T.; Liu, W.; Fukuyama, R.; Wang, Y.; Komori, H.; et al. Osteocalcin Is Necessary for the Alignment of Apatite Crystallites, but Not Glucose Metabolism, Testosterone Synthesis, or Muscle Mass. PLoS Genet. 2020, 16, e1008586. [Google Scholar] [CrossRef] [PubMed]
  24. Shan, C.; Ghosh, A.; Guo, X.; Wang, S.; Hou, Y.; Li, S.; Liu, J. Roles for Osteocalcin in Brain Signalling: Implications in Cognition- and Motor-Related Disorders. Mol. Brain 2019, 12, 23. [Google Scholar] [CrossRef]
  25. Si, J.; Wang, C.; Zhang, D.; Wang, B.; Hou, W.; Zhou, Y. Osteopontin in Bone Metabolism and Bone Diseases. Med. Sci. Monit. 2020, 26, e919159. [Google Scholar] [CrossRef]
  26. Tobeiha, M.; Moghadasian, M.H.; Amin, N.; Jafarnejad, S. RANKL/RANK/OPG Pathway: A Mechanism Involved in Exercise-Induced Bone Remodeling. BioMed Res. Int. 2020, 2020, 1–11. [Google Scholar] [CrossRef] [PubMed]
  27. Baud’huin, M.; Duplomb, L.; Teletchea, S.; Lamoureux, F.; Ruiz-Velasco, C.; Maillasson, M.; Redini, F.; Heymann, M.-F.; Heymann, D. Osteoprotegerin: Multiple Partners for Multiple Functions. Cytokine Growth Factor Rev. 2013, 24, 401–409. [Google Scholar] [CrossRef] [PubMed]
  28. Liu, Q.; Li, M.; Wang, S.; Xiao, Z.; Xiong, Y.; Wang, G. Recent Advances of Osterix Transcription Factor in Osteoblast Differentiation and Bone Formation. Front. Cell Dev. Biol. 2020, 8, 601224. [Google Scholar] [CrossRef]
  29. Komori, T. Regulation of Proliferation, Differentiation and Functions of Osteoblasts by Runx2. Int. J. Mol. Sci. 2019, 20, 1694. [Google Scholar] [CrossRef] [PubMed]
  30. Majidinia, M.; Sadeghpour, A.; Yousefi, B. The Roles of Signaling Pathways in Bone Repair and Regeneration. J. Cell Physiol. 2018, 233, 2937–2948. [Google Scholar] [CrossRef]
  31. Karner, C.M.; Long, F. Wnt Signaling and Cellular Metabolism in Osteoblasts. Cell. Mol. Life Sci. 2017, 74, 1649–1657. [Google Scholar] [CrossRef]
  32. Ballhause, T.M.; Jiang, S.; Baranowsky, A.; Brandt, S.; Mertens, P.R.; Frosch, K.-H.; Yorgan, T.; Keller, J. Relevance of Notch Signaling for Bone Metabolism and Regeneration. Int. J. Mol. Sci. 2021, 22, 1325. [Google Scholar] [CrossRef] [PubMed]
  33. Cao, X.; Chen, D. The BMP Signaling and in Vivo Bone Formation. Gene 2005, 357, 1–8. [Google Scholar] [CrossRef] [PubMed]
  34. Lowery, J.W.; Rosen, V. The BMP Pathway and Its Inhibitors in the Skeleton. Physiol. Rev. 2018, 98, 2431–2452. [Google Scholar] [CrossRef]
  35. Fang, H.; Zhu, D.; Yang, Q.; Chen, Y.; Zhang, C.; Gao, J.; Gao, Y. Emerging Zero-Dimensional to Four-Dimensional Biomaterials for Bone Regeneration. J. Nanobiotechnol. 2022, 20, 26. [Google Scholar] [CrossRef]
  36. Battafarano, G.; Rossi, M.; De Martino, V.; Marampon, F.; Borro, L.; Secinaro, A.; Del Fattore, A. Strategies for Bone Regeneration: From Graft to Tissue Engineering. Int. J. Mol. Sci. 2021, 22, 1128. [Google Scholar] [CrossRef]
  37. Girón, J.; Kerstner, E.; Medeiros, T.; Oliveira, L.; Machado, G.M.; Malfatti, C.F.; Pranke, P. Biomaterials for Bone Regeneration: An Orthopedic and Dentistry Overview. Braz. J. Med. Biol. Res. 2021, 54, e11055. [Google Scholar] [CrossRef]
  38. Qi, J.; Yu, T.; Hu, B.; Wu, H.; Ouyang, H. Current Biomaterial-Based Bone Tissue Engineering and Translational Medicine. Int. J. Mol. Sci. 2021, 22, 10233. [Google Scholar] [CrossRef]
  39. Guo, L.; Liang, Z.; Yang, L.; Du, W.; Yu, T.; Tang, H.; Li, C.; Qiu, H. The Role of Natural Polymers in Bone Tissue Engineering. J. Control. Release 2021, 338, 571–582. [Google Scholar] [CrossRef]
  40. Bharadwaz, A.; Jayasuriya, A.C. Recent Trends in the Application of Widely Used Natural and Synthetic Polymer Nanocomposites in Bone Tissue Regeneration. Mater. Sci. Eng. C 2020, 110, 110698. [Google Scholar] [CrossRef]
  41. Zhu, Y.; Goh, C.; Shrestha, A. Biomaterial Properties Modulating Bone Regeneration. Macromol. Biosci. 2021, 21, 2000365. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, L.; Yang, G.; Johnson, B.N.; Jia, X. Three-Dimensional (3D) Printed Scaffold and Material Selection for Bone Repair. Acta Biomater. 2019, 84, 16–33. [Google Scholar] [CrossRef]
  43. Su, X.; Xian, C.; Gao, M.; Liu, G.; Wu, J. Edible Materials in Tissue Regeneration. Macromol. Biosci. 2021, 21, 2100114. [Google Scholar] [CrossRef] [PubMed]
  44. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021, n71, e112. [Google Scholar] [CrossRef]
  45. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Moher, D. Updating Guidance for Reporting Systematic Reviews: Development of the PRISMA 2020 Statement. J. Clin. Epidemiol. 2021, 134, 103–112. [Google Scholar] [CrossRef]
  46. An, J.; Hao, D.; Zhang, Q.; Chen, B.; Zhang, R.; Wang, Y.; Yang, H. Natural Products for Treatment of Bone Erosive Diseases: The Effects and Mechanisms on Inhibiting Osteoclastogenesis and Bone Resorption. Int. Immunopharmacol. 2016, 36, 118–131. [Google Scholar] [CrossRef] [PubMed]
  47. Torre, E. Molecular Signaling Mechanisms behind Polyphenol-Induced Bone Anabolism. Phytochem. Rev. 2017, 16, 1183–1226. [Google Scholar] [CrossRef]
  48. Chircov, C.; Miclea, I.I.; Grumezescu, V.; Grumezescu, A.M. Essential Oils for Bone Repair and Regeneration—Mechanisms and Applications. Materials 2021, 14, 1867. [Google Scholar] [CrossRef]
  49. Schilling, T.; Ebert, R.; Raaijmakers, N.; Schütze, N.; Jakob, F. Effects of Phytoestrogens and Other Plant-Derived Compounds on Mesenchymal Stem Cells, Bone Maintenance and Regeneration. J. Steroid Biochem. Mol. Biol. 2014, 139, 252–261. [Google Scholar] [CrossRef]
  50. Singh, P.; Gupta, A.; Qayoom, I.; Singh, S.; Kumar, A. Orthobiologics with Phytobioactive Cues: A Paradigm in Bone Regeneration. Biomed. Pharmacother. 2020, 130, 110754. [Google Scholar] [CrossRef]
  51. Khosla, S.; Oursler, M.J.; Monroe, D.G. Estrogen and the Skeleton. Trends Endocrinol. Metab. 2012, 23, 576–581. [Google Scholar] [CrossRef]
  52. Domazetovic, V. Oxidative Stress in Bone Remodeling: Role of Antioxidants. CCMBM 2017, 14, 209. [Google Scholar] [CrossRef] [PubMed]
  53. Zhu, C.; Shen, S.; Zhang, S.; Huang, M.; Zhang, L.; Chen, X. Autophagy in Bone Remodeling: A Regulator of Oxidative Stress. Front. Endocrinol. 2022, 13, 898634. [Google Scholar] [CrossRef] [PubMed]
  54. Vuolteenaho, K.; Moilanen, T.; Moilanen, E. Non-Steroidal Anti-Inflammatory Drugs, Cyclooxygenase-2 and the Bone Healing Process. Basic Clin. Pharm. Toxicol 2007, 0, 149. [Google Scholar] [CrossRef]
  55. Schett, G. Effects of Inflammatory and Anti-Inflammatory Cytokines on the Bone: Cytokine Effects On Bone. Eur. J. Clin. Investig. 2011, 41, 1361–1366. [Google Scholar] [CrossRef] [PubMed]
  56. Dhawan, U.; Jaffery, H.; Salmeron-Sanchez, M.; Dalby, M.J. An Ossifying Landscape: Materials and Growth Factor Strategies for Osteogenic Signalling and Bone Regeneration. Curr. Opin. Biotechnol. 2022, 73, 355–363. [Google Scholar] [CrossRef] [PubMed]
  57. Han, Q.-Q.; Du, Y.; Yang, P.-S. The Role of Small Molecules in Bone Regeneration. Future Med. Chem. 2013, 5, 1671–1684. [Google Scholar] [CrossRef]
  58. Maeda, A. Recruitment of Mesenchymal Stem Cells to Damaged Sites by Plant-Derived Components. Front. Cell Dev. Biol. 2020, 8, 437. [Google Scholar] [CrossRef]
  59. Kornicka, K.; Kocherova, I.; Marycz, K. The Effects of Chosen Plant Extracts and Compounds on Mesenchymal Stem Cells-a Bridge between Molecular Nutrition and Regenerative Medicine- Concise Review. Phytother. Res. 2017, 31, 947–958. [Google Scholar] [CrossRef]
  60. Mao, W.; Huang, G.; Chen, H.; Xu, L.; Qin, S.; Li, A. Research Progress of the Role of Anthocyanins on Bone Regeneration. Front. Pharm. 2021, 12, 773660. [Google Scholar] [CrossRef]
  61. Martiniakova, M.; Babikova, M.; Omelka, R. Pharmacological Agents and Natural Compounds: Available Treatments for Osteoporosis. J. Physiol. Pharm. 2020, 71, 1–4. [Google Scholar] [CrossRef]
  62. Sarkar, N.; Bose, S. Controlled Release of Soy Isoflavones from Multifunctional 3D Printed Bone Tissue Engineering Scaffolds. Acta Biomater. 2020, 114, 407–420. [Google Scholar] [CrossRef] [PubMed]
  63. Xu, Z.; Sun, Y.; Dai, H.; Ma, Y.; Bing, H. Engineered 3D-Printed Polyvinyl Alcohol Scaffolds Incorporating β-Tricalcium Phosphate and Icariin Induce Bone Regeneration in Rat Skull Defect Model. Molecules 2022, 27, 4535. [Google Scholar] [CrossRef]
  64. Burim, R.A.; Sendyk, D.I.; Hernandes, L.S.; de Souza, D.F.M.; Correa, L.; Deboni, M.C.Z. Repair of Critical Calvarias Defects With Systemic Epimedium Sagittatum Extract. J. Craniofac. Surg. 2016, 27, 799–804. [Google Scholar] [CrossRef] [PubMed]
  65. You, M.; Jing, J.; Tian, D.; Qian, J.; Yu, G. Dioscin Stimulates Differentiation of Mesenchymal Stem Cells towards Hypertrophic Chondrocytes in Vitro and Endochondral Ossification in Vivo. Am J Transl Res 2016, 8, 3930–3938. [Google Scholar]
  66. Adhikary, S.; Choudhary, D.; Ahmad, N.; Karvande, A.; Kumar, A.; Banala, V.T.; Mishra, P.R.; Trivedi, R. Dietary Flavonoid Kaempferol Inhibits Glucocorticoid-Induced Bone Loss by Promoting Osteoblast Survival. Nutrition 2018, 53, 64–76. [Google Scholar] [CrossRef]
  67. Zhao, J.; Wu, J.; Xu, B.; Yuan, Z.; Leng, Y.; Min, J.; Lan, X.; Luo, J. Kaempferol Promotes Bone Formation in Part via the MTOR Signaling Pathway. Mol. Med. Rep. 2019, 20, 5197–5207. [Google Scholar] [CrossRef]
  68. Kulkarni, C.; Sharma, S.; Bora, P.S.; Verma, S.; Rajput, S.; Porwal, K.; Rath, S.K.; Gayen, J.R.; Sharma, U.; Chattopadhyay, N. A Novel Extraction Method Enhanced the Osteogenic and Anti-Osteoporosis Effect of Tea Extract without Any Hepatotoxicity in Ovariectomized Rats. Front. Endocrinol. 2022, 13. [Google Scholar] [CrossRef]
  69. Wei, Y.; Fu, J.; Wu, W.; Ma, P.; Ren, L.; Yi, Z.; Wu, J. Quercetin Prevents Oxidative Stress-Induced Injury of Periodontal Ligament Cells and Alveolar Bone Loss in Periodontitis. Drug. Des. Devel. 2021, 15, 3509–3522. [Google Scholar] [CrossRef] [PubMed]
  70. Khedgikar, V.; Kushwaha, P.; Ahmad, N.; Gautam, J.; Kumar, P.; Maurya, R.; Trivedi, R. Ethanolic Extract of Dalbergia Sissoo Promotes Rapid Regeneration of Cortical Bone in Drill-Hole Defect Model of Rat. Biomed Pharm. 2017, 86, 16–22. [Google Scholar] [CrossRef]
  71. Gong, W.; Zhang, N.; Cheng, G.; Zhang, Q.; He, Y.; Shen, Y.; Zhang, Q.; Zhu, B.; Zhang, Q.; Qin, L. Rehmannia Glutinosa Libosch Extracts Prevent Bone Loss and Architectural Deterioration and Enhance Osteoblastic Bone Formation by Regulating the IGF-1/PI3K/MTOR Pathway in Streptozotocin-Induced Diabetic Rats. Int. J. Mol. Sci. 2019, 20, 3964. [Google Scholar] [CrossRef]
  72. Tan, B.; Wu, Y.; Wu, Y.; Shi, K.; Han, R.; Li, Y.; Qian, Z.; Liao, J. Curcumin-Microsphere/IR820 Hybrid Bifunctional Hydrogels for In Situ Osteosarcoma Chemo-Co-Thermal Therapy and Bone Reconstruction. ACS Appl. Mater. Interfaces 2021, 13, 31542–31553. [Google Scholar] [CrossRef] [PubMed]
  73. Verma, A.H.; Kumar, T.S.S.; Madhumathi, K.; Rubaiya, Y.; Ramalingan, M.; Doble, M. Curcumin Releasing Eggshell Derived Carbonated Apatite Nanocarriers for Combined Anti-Cancer, Anti-Inflammatory and Bone Regenerative Therapy. J. Nanosci. Nanotechnol. 2019, 19, 6872–6880. [Google Scholar] [CrossRef] [PubMed]
  74. Li, Y.; Dånmark, S.; Edlund, U.; Finne-Wistrand, A.; He, X.; Norgård, M.; Blomén, E.; Hultenby, K.; Andersson, G.; Lindgren, U. Resveratrol-Conjugated Poly-ε-Caprolactone Facilitates in vitro Mineralization and in vivo Bone Regeneration. Acta Biomater. 2011, 7, 751–758. [Google Scholar] [CrossRef] [PubMed]
  75. Murgia, D.; Mauceri, R.; Campisi, G.; De Caro, V. Advance on Resveratrol Application in Bone Regeneration: Progress and Perspectives for Use in Oral and Maxillofacial Surgery. Biomolecules 2019, 9, 94. [Google Scholar] [CrossRef]
  76. Bhattarai, G.; Poudel, S.B.; Kook, S.-H.; Lee, J.-C. Resveratrol Prevents Alveolar Bone Loss in an Experimental Rat Model of Periodontitis. Acta Biomater. 2016, 29, 398–408. [Google Scholar] [CrossRef]
  77. Choi, Y.; Yoon, D.S.; Lee, K.-M.; Choi, S.M.; Lee, M.-H.; Park, K.H.; Han, S.H.; Lee, J.W. Enhancement of Mesenchymal Stem Cell-Driven Bone Regeneration by Resveratrol-Mediated SOX2 Regulation. Aging Dis. 2019, 10, 818–833. [Google Scholar] [CrossRef]
  78. Kim, J.-S.; Takanche, J.S.; Kim, J.-E.; Jeong, S.-H.; Han, S.-H.; Yi, H.-K. Schisandra Chinensis Extract Ameliorates Age-Related Muscle Wasting and Bone Loss in Ovariectomized Rats. Phytother. Res. 2019, 33, 1865–1877. [Google Scholar] [CrossRef]
  79. Suliman, S.; Mieszkowska, A.; Folkert, J.; Rana, N.; Mohamed-Ahmed, S.; Fuoco, T.; Finne-Wistrand, A.; Dirscherl, K.; Jørgensen, B.; Mustafa, K.; et al. Immune-Instructive Copolymer Scaffolds Using Plant-Derived Nanoparticles to Promote Bone Regeneration. Inflamm. Regen. 2022, 42, 12. [Google Scholar] [CrossRef]
  80. Folkert, J.; Meresta, A.; Gaber, T.; Miksch, K.; Buttgereit, F.; Detert, J.; Pischon, N.; Gurzawska, K. Nanocoating with Plant-Derived Pectins Activates Osteoblast Response in vitro. Int. J. Nanomed. 2016, 12, 239–249. [Google Scholar] [CrossRef]
  81. Soares, I.M.V.; Fernandes, G.V. de O.; Cavalcante, L.C.; Leite, Y.K.P. de C.; Bezerra, D. de O.; de Carvalho, M.A.M.; Carvalho, C.M.R.S. 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. J. Appl Oral Sci. 2019, 27, e20180103. [Google Scholar] [CrossRef]
  82. Lin, Z.; Lin, C.; Fu, C.; Lu, H.; Jin, H.; Chen, Q.; Pan, J. The Protective Effect of Ellagic Acid (EA) in Osteoarthritis: An in vitro and in Vivo Study. Biomed. Pharm. 2020, 125, 109845. [Google Scholar] [CrossRef] [PubMed]
  83. Fikry, E.M.; Gad, A.M.; Eid, A.H.; Arab, H.H. Caffeic Acid and Ellagic Acid Ameliorate Adjuvant-Induced Arthritis in Rats via Targeting Inflammatory Signals, Chitinase-3-like Protein-1 and Angiogenesis. Biomed Pharm. 2019, 110, 878–886. [Google Scholar] [CrossRef] [PubMed]
  84. Ding, L.; Gu, S.; Zhou, B.; Wang, M.; Zhang, Y.; Wu, S.; Zou, H.; Zhao, G.; Gao, Z.; Xu, L. Ginsenoside Compound K Enhances Fracture Healing via Promoting Osteogenesis and Angiogenesis. Front Pharm. 2022, 13, 855393. [Google Scholar] [CrossRef]
  85. Zhang, Y.; Ma, J.; Zhang, W. Berberine for Bone Regeneration: Therapeutic Potential and Molecular Mechanisms. J. Ethnopharmacol. 2021, 277, 114249. [Google Scholar] [CrossRef]
  86. Dong, B.; Liu, X.; Li, J.; Wang, B.; Yin, J.; Zhang, H.; Liu, W. Berberine Encapsulated in Exosomes Derived from Platelet-Rich Plasma Promotes Chondrogenic Differentiation of the Bone Marrow Mesenchymal Stem Cells via the Wnt/β-Catenin Pathway. Biol. Pharm. Bull. 2022, 45, 1444–1451. [Google Scholar] [CrossRef]
  87. Shaban, N.Z.; Kenawy, M.Y.; Taha, N.A.; Abd El-Latif, M.M.; Ghareeb, D.A. Cellulose Acetate Nanofibers: Incorporating Hydroxyapatite (HA), HA/Berberine or HA/Moghat Composites, as Scaffolds to Enhance In vitro Osteoporotic Bone Regeneration. Polymers 2021, 13, 4140. [Google Scholar] [CrossRef]
  88. Zhang, R.; Yang, J.; Wu, J.; Xiao, L.; Miao, L.; Qi, X.; Li, Y.; Sun, W. Berberine Promotes Osteogenic Differentiation of Mesenchymal Stem Cells with Therapeutic Potential in Periodontal Regeneration. Eur. J. Pharmacol. 2019, 851, 144–150. [Google Scholar] [CrossRef]
  89. Talebi, A.; Hayati Roodbari, N.; Reza Sameni, H.; Zarbakhsh, S. Impact of Coadministration of Apigenin and Bone Marrow Stromal Cells on Damaged Ovaries Due to Chemotherapy in Rat: An Experimental Study. Int. J. Reprod. Biomed. 2020, 18, 551–560. [Google Scholar] [CrossRef]
  90. Mao, Y.-W.; Lin, R.-D.; Hung, H.-C.; Lee, M.-H. Stimulation of Osteogenic Activity in Human Osteoblast Cells by Edible Uraria Crinita. J. Agric. Food Chem. 2014, 62, 5581–5588. [Google Scholar] [CrossRef]
  91. Pal, S.; Kumar, P.; Ramakrishna, E.; Kumar, S.; Porwal, K.; Kumar, B.; Arya, K.R.; Maurya, R.; Chattopadhyay, N. Extract and Fraction of Cassia Occidentalis L. (a Synonym of Senna Occidentalis) Have Osteogenic Effect and Prevent Glucocorticoid-Induced Osteopenia. J. Ethnopharmacol. 2019, 235, 8–18. [Google Scholar] [CrossRef] [PubMed]
  92. Pal, S.; Mittapelly, N.; Husain, A.; Kushwaha, S.; Chattopadhyay, S.; Kumar, P.; Ramakrishna, E.; Kumar, S.; Maurya, R.; Sanyal, S.; et al. A Butanolic Fraction from the Standardized Stem Extract of Cassia Occidentalis L Delivered by a Self-Emulsifying Drug Delivery System Protects Rats from Glucocorticoid-Induced Osteopenia and Muscle Atrophy. Sci. Rep. 2020, 10, 195. [Google Scholar] [CrossRef]
  93. Zhou, R.P.; Lin, S.J.; Wan, W.B.; Zuo, H.L.; Yao, F.F.; Ruan, H.B.; Xu, J.; Song, W.; Zhou, Y.C.; Wen, S.Y.; et al. Chlorogenic Acid Prevents Osteoporosis by Shp2/PI3K/Akt Pathway in Ovariectomized Rats. PLoS ONE 2016, 11, e0166751. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, M.; Hu, X. Mechanism of Chlorogenic Acid Treatment on Femoral Head Necrosis and Its Protection of Osteoblasts. Biomed. Rep. 2016, 5, 57–62. [Google Scholar] [CrossRef]
  95. Na, W.; Kang, M.-K.; Park, S.-H.; Kim, D.Y.; Oh, S.Y.; Oh, M.-S.; Park, S.; Kang, I.-J.; Kang, Y.-H. Aesculetin Accelerates Osteoblast Differentiation and Matrix-Vesicle-Mediated Mineralization. Int. J. Mol. Sci. 2021, 22, 12391. [Google Scholar] [CrossRef]
  96. Kurian, I.G.; Dileep, P.; Ipshita, S.; Pradeep, A.R. Comparative Evaluation of Subgingivally-Delivered 1% Metformin and Aloe Vera Gel in the Treatment of Intrabony Defects in Chronic Periodontitis Patients: A Randomized, Controlled Clinical Trial. J Investig. Clin. Dent. 2018, 9, e12324. [Google Scholar] [CrossRef]
  97. Tanık, A.; Güler Doğru, A.; Akpolat, V.; Acun Kaya, F.; Sarıbaş, E.; Gül, M.; İrtegün Kandemir, S.; Deveci, E. Investigation of the Effect of Combined Use of Alloplastic-Based Tricalcium Phosphate Bone Graft and Antihemorrhagic Plant Extract (ABS) on Bone Regeneration in Surgically Induced Bone Defects in Nondiabetic Rats: An Experimental Animal Study. Turk. J. Med. Sci. 2018, 48, 1302–1314. [Google Scholar] [CrossRef] [PubMed]
  98. Khedgikar, V.; Kushwaha, P.; Gautam, J.; Verma, A.; Changkija, B.; Kumar, A.; Sharma, S.; Nagar, G.K.; Singh, D.; Trivedi, P.K.; et al. Withaferin A: A Proteasomal Inhibitor Promotes Healing after Injury and Exerts Anabolic Effect on Osteoporotic Bone. Cell Death Dis. 2013, 4, e778. [Google Scholar] [CrossRef]
  99. Yan, C.-P.; Wang, X.-K.; Jiang, K.; Yin, C.; Xiang, C.; Wang, Y.; Pu, C.; Chen, L.; Li, Y.-L. β-Ecdysterone Enhanced Bone Regeneration through the BMP-2/SMAD/RUNX2/Osterix Signaling Pathway. Front. Cell Dev. Biol. 2022, 10. [Google Scholar] [CrossRef]
  100. Li, F.; Yang, X.; Yang, Y.; Guo, C.; Zhang, C.; Yang, Z.; Li, P. Antiosteoporotic Activity of Echinacoside in Ovariectomized Rats. Phytomedicine 2013, 20, 549–557. [Google Scholar] [CrossRef] [PubMed]
  101. Madhurakkat, P.S.K.; Lee, S.M.; Lee, J.; Ahmad, T.; Lee, M.S.; Yang, H.S.; Shin, H. Oxidative Epigallocatechin Gallate Coating on Polymeric Substrates for Bone Tissue Regeneration. Macromol. Biosci. 2019, 19, e1800392. [Google Scholar] [CrossRef]
  102. Sasayama, S.; Hara, T.; Tanaka, T.; Honda, Y.; Baba, S. Osteogenesis of Multipotent Progenitor Cells Using the Epigallocatechin Gallate-Modified Gelatin Sponge Scaffold in the Rat Congenital Cleft-Jaw Model. Int. J. Mol. Sci. 2018, 19, 3803. [Google Scholar] [CrossRef]
  103. Naumenko, E.; Guryanov, I.; Zakirova, E.; Fakhrullin, R. Forskolin-Loaded Halloysite Nanotubes as Osteoconductive Additive for the Biopolymer Tissue Engineering Scaffolds. Polymers 2021, 13, 3949. [Google Scholar] [CrossRef]
  104. Douglas, T.E.L.; Keppler, J.K.; Vandrovcová, M.; Plencner, M.; Beranová, J.; Feuereisen, M.; Parakhonskiy, B.V.; Svenskaya, Y.; Atkin, V.; Ivanova, A.; et al. Enhancement of Biomimetic Enzymatic Mineralization of Gellan Gum Polysaccharide Hydrogels by Plant-Derived Gallotannins. Int. J. Mol. Sci. 2020, 21, 2315. [Google Scholar] [CrossRef]
  105. Porwal, K.; Pal, S.; Dev, K.; China, S.P.; Kumar, Y.; Singh, C.; Barbhuyan, T.; Sinha, N.; Sanyal, S.; Trivedi, A.K.; et al. Guava Fruit Extract and Its Triterpene Constituents Have Osteoanabolic Effect: Stimulation of Osteoblast Differentiation by Activation of Mitochondrial Respiration via the Wnt/β-Catenin Signaling. J. Nutr. Biochem. 2017, 44, 22–34. [Google Scholar] [CrossRef] [PubMed]
  106. Hu, B.; Chen, L.; Chen, Y.; Zhang, Z.; Wang, X.; Zhou, B. Cyanidin-3-Glucoside Regulates Osteoblast Differentiation via the ERK1/2 Signaling Pathway. ACS Omega 2021, 6, 4759–4766. [Google Scholar] [CrossRef] [PubMed]
  107. Gu, H.; Boonanantanasarn, K.; Kang, M.; Kim, I.; Woo, K.M.; Ryoo, H.-M.; Baek, J.-H. Morinda Citrifolia Leaf Extract Enhances Osteogenic Differentiation Through Activation of Wnt/β-Catenin Signaling. J. Med. Food. 2018, 21, 57–69. [Google Scholar] [CrossRef]
  108. Boonanantanasarn, K.; Janebodin, K.; Suppakpatana, P.; Arayapisit, T.; Rodsutthi, J.; Chunhabundit, P.; Boonanuntanasarn, S.; Sripairojthikoon, W. Morinda Citrifolia Leaves Enhance Osteogenic Differentiation and Mineralization of Human Periodontal Ligament Cells. Dent. Mater. J. 2014, 33, 157–165. [Google Scholar] [CrossRef]
  109. Shalan, N.A.A.M.; Mustapha, N.M.; Mohamed, S. Noni Leaf and Black Tea Enhance Bone Regeneration in Estrogen-Deficient Rats. Nutrition 2017, 33, 42–51. [Google Scholar] [CrossRef]
  110. Kalalinia, F.; Ghasim, H.; Amel Farzad, S.; Pishavar, E.; Ramezani, M.; Hashemi, M. Comparison of the Effect of Crocin and Crocetin, Two Major Compounds Extracted from Saffron, on Osteogenic Differentiation of Mesenchymal Stem Cells. Life Sci. 2018, 208, 262–267. [Google Scholar] [CrossRef] [PubMed]
  111. Kim, S.S.; Kim, N.K.; Seo, S.R. Cynanchi Atrati and Its Phenolic Constituent Sinapic Acid Target Regulator of Calcineurin 1 (RCAN1) to Control Skin Inflammation. Antioxid 2022, 11, 205. [Google Scholar] [CrossRef]
  112. Balagangadharan, K.; Trivedi, R.; Vairamani, M.; Selvamurugan, N. Sinapic Acid-Loaded Chitosan Nanoparticles in Polycaprolactone Electrospun Fibers for Bone Regeneration in vitro and in vivo. Carbohydr. Polym. 2019, 216, 1–16. [Google Scholar] [CrossRef]
  113. Abiramasundari, G.; Gowda, C.M.M.; Pampapathi, G.; Praveen, S.; Shivamurugan, S.; Vijaykumar, M.; Devi, A.; Sreepriya, M. Ethnomedicine Based Evaluation of Osteoprotective Properties of Tinospora Cordifolia on in Vitro and in Vivo Model Systems. Biomed. Pharm. 2017, 87, 342–354. [Google Scholar] [CrossRef] [PubMed]
  114. Cheng, W.-X.; Liu, Y.-Z.; Meng, X.-B.; Zheng, Z.-T.; Li, L.-L.; Ke, L.-Q.; Li, L.; Huang, C.-S.; Zhu, G.-Y.; Pan, H.-D.; et al. PLGA/β-TCP Composite Scaffold Incorporating Cucurbitacin B Promotes Bone Regeneration by Inducing Angiogenesis. J. Orthop. Transl. 2021, 31, 41–51. [Google Scholar] [CrossRef] [PubMed]
  115. Xie, Y.; Jiang, Z.; Yang, R.; Ye, Y.; Pei, L.; Xiong, S.; Wang, S.; Wang, L.; Liu, S. Polysaccharide-Rich Extract from Polygonatum Sibiricum Protects Hematopoiesis in Bone Marrow Suppressed by Triple Negative Breast Cancer. Biomed. Pharm. 2021, 137, 111338. [Google Scholar] [CrossRef] [PubMed]
  116. Wardhana, A.S.; Nirwana, I.; Budi, H.S.; Surboyo, M.D.C. Role of Hydroxyapatite and Ellagic Acid in the Osteogenesis. Eur. J. Dent. 2021, 15, 8–12. [Google Scholar] [CrossRef]
  117. Xu, W.; Xu, J.; Wang, T.; Liu, W.; Wei, H.; Yang, X.; Yan, W.; Zhou, W.; Xiao, J. Ellagic Acid and Sennoside B Inhibit Osteosarcoma Cell Migration, Invasion and Growth by Repressing the Expression of c-Jun. Oncol. Lett. 2018, 16, 898–904. [Google Scholar] [CrossRef]
  118. Nirwana, I.; Munadziroh, E.; Yuliati, A.; Fadhila, A.I.; Nurliana; Wardhana, A.S.; Shariff, K.A.; Surboyo, M.D.C. Ellagic Acid and Hydroxyapatite Promote Angiogenesis Marker in Bone Defect. J. Oral Biol. Craniofac. Res. 2022, 12, 116–120. [Google Scholar] [CrossRef]
  119. Primasari, D.N.; Nirwana, I.; Budi, H.S.; Wardhana, A.S.; Sari, A.F.; Novita, N.; Setyawan, A.P.; Surboyo, M.D.C.; Shariff, K.A. The Cytokine and Bone Protein Expression by Ellagic Acid-Hydroxyapatite in Bone Remodelling Model. Sci. World J. 2022, 2022, 6740853. [Google Scholar] [CrossRef]
  120. Pang, X.; Zhong, Z.; Jiang, F.; Yang, J.; Nie, H. Juglans Regia L. Extract Promotes Osteogenesis of Human Bone Marrow Mesenchymal Stem Cells through BMP2/Smad/Runx2 and Wnt/β-Catenin Pathways. J. Orthop. Surg. Res. 2022, 17, 88. [Google Scholar] [CrossRef]
  121. Vandenbroucke, A.; Luyten, F.P.; Flamaing, J.; Gielen, E. Pharmacological Treatment of Osteoporosis in the Oldest Old. Clin Interv. Aging 2017, 12, 1065–1077. [Google Scholar] [CrossRef]
  122. Brown, J.P. Long-Term Treatment of Postmenopausal Osteoporosis. Endocrinol. Metab. 2021, 36, 544–552. [Google Scholar] [CrossRef] [PubMed]
  123. Du, Z.; Wang, C.; Zhang, R.; Wang, X.; Li, X. Applications of Graphene and Its Derivatives in Bone Repair: Advantages for Promoting Bone Formation and Providing Real-Time Detection, Challenges and Future Prospects. Int. J. Nanomed. 2020, 15, 7523–7551. [Google Scholar] [CrossRef] [PubMed]
  124. Masne, N.; Ambade, R.; Bhugaonkar, K. Use of Nanocomposites in Bone Regeneration. Cureus 2022, 14, e31346. [Google Scholar] [CrossRef]
  125. Qaseem, A.; Forciea, M.A.; McLean, R.M.; Denberg, T.D.; Clinical Guidelines Committee of the American College of Physicians; Barry, M.J.; Cooke, M.; Fitterman, N.; Harris, R.P.; Humphrey, L.L.; et al. Treatment of Low Bone Density or Osteoporosis to Prevent Fractures in Men and Women: A Clinical Practice Guideline Update From the American College of Physicians. Ann. Intern. Med. 2017, 166, 818–839. [Google Scholar] [CrossRef] [PubMed]
  126. An, J.; Yang, H.; Zhang, Q.; Liu, C.; Zhao, J.; Zhang, L.; Chen, B. Natural Products for Treatment of Osteoporosis: The Effects and Mechanisms on Promoting Osteoblast-Mediated Bone Formation. Life Sci. 2016, 147, 46–58. [Google Scholar] [CrossRef]
  127. Raut, N.; Wicks, S.M.; Lawal, T.O.; Mahady, G.B. Epigenetic Regulation of Bone Remodeling by Natural Compounds. Pharm. Res. 2019, 147, 104350. [Google Scholar] [CrossRef]
  128. Shanmugavadivu, A.; Balagangadharan, K.; Selvamurugan, N. Angiogenic and Osteogenic Effects of Flavonoids in Bone Regeneration. Biotechnol. Bioeng. 2022, 119, 2313–2330. [Google Scholar] [CrossRef]
  129. Qiao, K.; Xu, L.; Tang, J.; Wang, Q.; Lim, K.S.; Hooper, G.; Woodfield, T.B.F.; Liu, G.; Tian, K.; Zhang, W.; et al. The Advances in Nanomedicine for Bone and Cartilage Repair. J. Nanobiotechnology 2022, 20, 141. [Google Scholar] [CrossRef]
  130. Jin, G.-Z. Current Nanoparticle-Based Technologies for Osteoarthritis Therapy. Nanomaterials 2020, 10, 2368. [Google Scholar] [CrossRef]
  131. Mu, P.; Feng, J.; Hu, Y.; Xiong, F.; Ma, X.; Tian, L. Botanical Drug Extracts Combined With Biomaterial Carriers for Osteoarthritis Cartilage Degeneration Treatment: A Review of 10 Years of Research. Front. Pharm. 2021, 12, 789311. [Google Scholar] [CrossRef]
  132. Song, H.; Li, X.; Zhao, Z.; Qian, J.; Wang, Y.; Cui, J.; Weng, W.; Cao, L.; Chen, X.; Hu, Y.; et al. Reversal of Osteoporotic Activity by Endothelial Cell-Secreted Bone Targeting and Biocompatible Exosomes. Nano Lett. 2019, 19, 3040–3048. [Google Scholar] [CrossRef] [PubMed]
  133. Paesa, M.; Alejo, T.; Garcia-Alvarez, F.; Arruebo, M.; Mendoza, G. New Insights in Osteoarthritis Diagnosis and Treatment: Nano-Strategies for an Improved Disease Management. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2023, 15, e1844. [Google Scholar] [CrossRef] [PubMed]
  134. Gao, Z.-R.; Feng, Y.-Z.; Zhao, Y.-Q.; Zhao, J.; Zhou, Y.-H.; Ye, Q.; Chen, Y.; Tan, L.; Zhang, S.-H.; Feng, Y.; et al. Traditional Chinese Medicine Promotes Bone Regeneration in Bone Tissue Engineering. Chin Med 2022, 17, 86. [Google Scholar] [CrossRef] [PubMed]
  135. Bose, S.; Sarkar, N.; Banerjee, D. Natural Medicine Delivery from Biomedical Devices to Treat Bone Disorders: A Review. Acta Biomater. 2021, 126, 63–91. [Google Scholar] [CrossRef] [PubMed]
  136. Cavalu, S.; Simon, V.; Goller, G.; Akin, I. Bioactivity And Antimicrobial Properties Of Pmma/Ag2o Acrylic Bone Cement Collagen Coated. Dig. J. Nanomater. Biostruct. 2011, 6, 779–790. [Google Scholar]
  137. Zhang, X.; Li, Z.; Yang, P.; Duan, G.; Liu, X.; Gu, Z.; Li, Y. Polyphenol Scaffolds in Tissue Engineering. Mater. Horiz. 2021, 8, 145–167. [Google Scholar] [CrossRef] [PubMed]
  138. Monárrez-Cordero, B.E.; Rodríguez-González, C.A.; Valencia-Gómez, L.E.; Hernández-Paz, J.F.; Martel-Estrada, S.A.; Camacho-Montes, H.; Olivas-Armendáriz, I. The Effect of Allium Cepa Extract on the Chitosan/PLGA Scaffolds Bioactivity. J. Appl. Biomater. Funct. Mater. 2021, 19, 2280800021989701. [Google Scholar] [CrossRef]
  139. Tahmasebi, A.; Shapouri Moghadam, A.; Enderami, S.E.; Islami, M.; Kaabi, M.; Saburi, E.; Daei Farshchi, A.; Soleimanifar, F.; Mansouri, V. Aloe Vera-Derived Gel-Blended PHBV Nanofibrous Scaffold for Bone Tissue Engineering. ASAIO J. 2020, 66, 966–973. [Google Scholar] [CrossRef]
  140. Shanmugavel, S.; Reddy, V.J.; Ramakrishna, S.; Lakshmi, B.; Dev, V.G. Precipitation of Hydroxyapatite on Electrospun Polycaprolactone/Aloe Vera/Silk Fibroin Nanofibrous Scaffolds for Bone Tissue Engineering. J. Biomater. Appl. 2014, 29, 46–58. [Google Scholar] [CrossRef]
  141. Mohammadpour, M.; Samadian, H.; Moradi, N.; Izadi, Z.; Eftekhari, M.; Hamidi, M.; Shavandi, A.; Quéro, A.; Petit, E.; Delattre, C.; et al. Fabrication and Characterization of Nanocomposite Hydrogel Based on Alginate/Nano-Hydroxyapatite Loaded with Linum Usitatissimum Extract as a Bone Tissue Engineering Scaffold. Mar. Drugs 2021, 20, 20. [Google Scholar] [CrossRef]
  142. Garcia, C.F.; Marangon, C.A.; Massimino, L.C.; Klingbeil, M.F.G.; Martins, V.C.A.; de Guzzi Plepis, A.M. Development of Collagen/Nanohydroxyapatite Scaffolds Containing Plant Extract Intended for Bone Regeneration. Mater. Sci. Eng. C 2021, 123, 111955. [Google Scholar] [CrossRef] [PubMed]
  143. Wang, Z.; Liu, H.; Luo, W.; Cai, T.; Li, Z.; Liu, Y.; Gao, W.; Wan, Q.; Wang, X.; Wang, J.; et al. Regeneration of Skeletal System with Genipin Crosslinked Biomaterials. J. Tissue Eng. 2020, 11, 2041731420974861. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Prisma flow-diagram for description of the selection process of the bibliographic sources.
Figure 1. Prisma flow-diagram for description of the selection process of the bibliographic sources.
Plants 12 02055 g001
Figure 2. The main classes of phytocompounds and the main regulatory mechanisms involved in the bone regeneration process, the stages of bone density restoration in different pathologies.
Figure 2. The main classes of phytocompounds and the main regulatory mechanisms involved in the bone regeneration process, the stages of bone density restoration in different pathologies.
Plants 12 02055 g002
Figure 3. The diversity of nanometric systems used for the innovative application of phytocompounds for the purpose of bone regeneration.
Figure 3. The diversity of nanometric systems used for the innovative application of phytocompounds for the purpose of bone regeneration.
Plants 12 02055 g003
Table 1. Phytochemical compounds involved in bone regeneration process including their mechanism of action and natural sources.
Table 1. Phytochemical compounds involved in bone regeneration process including their mechanism of action and natural sources.
CompoundsType of
Activity
Mechanism of
Action
Extract
Source
In vivo/in vitro StudiesRef.
Genistein
Plants 12 02055 i001
Proestrogenic
activity
↑ alkaline phosphatase level
↓ urinary excretion of calcium and phosphate, → serum concentration at the appropriate normal level
Erythrina variegateIn vitro (mesenchymal stem cells)[61]
Daidzein
Plants 12 02055 i002
↑ osteoclast apoptosis through the mediation of estrogen receptors
↓ the loss of bone density
activates tyrosine phosphatase → ↓ membrane depolarization producing changes in intracellular Ca2+
In vivo (rats)[62]
Icariin
Plants 12 02055 i003
↓ bone loss in the median bone area by regulating the ratio between osteoprotegerin and RANKL, which are key mediators of osteoclast genesis.
↑ proliferation, differentiation of osteoblasts, bone mineralization
↓ cell apoptosis
direct osteoblast stimulation: activation of the bone morphogenetic protein (BMP) cascade through (promoting Runx2/Cbfa1 expression and the production of BMP-4, BMP-2, and SMAD4 and nitrous oxide release; high levels of ALP
suppression of p38 and JNK pathways in the osteoclasts, ↓ release of prostaglandin E2 by osteoblasts => inhibition of osteoclast differentiation





Epimedium




In vivo (rats)




[63]



[64]
Dioscine
Plants 12 02055 i004
↑the proliferation of bone tissue
↓ cell apoptosis by mediating signaling pathways
↓RANKL expression
↓osteoprotegerin/RANKL → inhibits bone reabsorption
Dioscoreaceae familyIn vivo (mice)[65]

Kaempferol
Plants 12 02055 i005
↑ osteoprotegerin and ↓ RANKL expression → osteoclastogenesis decreases
↑ antiapoptotic expression
maintaining bone mass, microarchitecture, and bone strength of the trabecular bones
Ginkgo biloba



Camellia sinensis
In vivo (rats)
In vivo (rats)


In vivo (rats)
[66,67,68]


[69]
Quercetin
Plants 12 02055 i006
↑ the proliferation of bone tissue
↓osteoprotegerin/RANKL → inhibits bone reabsorption
In vitro (mesenchymal stem cells)

In vitro (periodontal ligament cells)
[61,69]
Ginkgolic acid
Plants 12 02055 i007
↑ proliferation, differentiation of osteoblasts, bone mineralizationGinkgo biloba
In vitro (mesenchymal stem cells)





[61]



Caviunin
Plants 12 02055 i008
stimulates BMP-2/Wnt-βcatenin pathwayDalbergia sissooIn vivo (rats)[70]
Acteoside
Plants 12 02055 i009
Antioxidant and anti-inflammatory effect↓ the level of pro-inflammatory cytokines such as TNF-α and IL-6,
↓ the differentiation of osteoclasts by reducing free radicals and fighting oxidative stress
↑ cell proliferation
↓ bone demineralization
Verbascum sp.
Cistanche sp.
In vivo (rats)[71]
Curcumin
Plants 12 02055 i010
↓the level of inflammation by decreasing the inflammatory cytokines TNF-a and IL-6
↓bone loss and demineralization, inhibiting osteoclastogenesis
↑the level of alkaline phosphatase, which leads to an increase in the mineralization process

interaction with transcription and growth factors, protein kinases, cytokines and enzymes => apoptosis of cancer cell
Curcuma longaIn vitro (osteosarcoma cells)


In vitro (human osteosarcoma cells)
[72]



[73]
Resveratrol
Plants 12 02055 i011
↓the level of free radicals from the bone level, neutralizing them
↓bone loss
inhibits osteoclastogenesis and the RANKL marker
influences the response of estrogen receptors to oxidative stress factors
↑bone differentiation → ↑ bone density
↑the level of morphogenetic protein at the bone level
↓decreases the level of alkaline phosphatase
↓the level of osteocalcin.

allows mass production of MSCs; mRNA levels of RUNX2, Collagen Type I Alpha 1 (COL1A1), PPARγ, Adiponectin (APN) were highly expressed,
↑ SIRT1 and SOX2 levels
-








In vivo (rats)
In vivo (rats)






In vitro (mesenchymal stem cells)







[74,75,76,77]







[77]
Gomisin
Plants 12 02055 i012




Schisandrin C
Plants 12 02055 i013
down-regulation of inflammatory molecules, ROS, and up-regulation of antioxidant moleculesSchisandra chinensisIn vitro (murine macrophage, myoblasts, human diploid fibroblasts, bone marrow macrophages, osteoblasts)
In vivo (rats)
[78]
Rhamnogalacturonan-IPlants 12 02055 i014↓ intracellular accumulation of galectin-3
down-regulation of RANKL, TNFα, IL-6, and IL-1β
Solanum tuberosumIn vitro (neutrophils and macrophages; osteoblasts)
In vivo (rats)
[79,80]
Acemannan
Plants 12 02055 i015
tissue regeneration, cell proliferation, extracellular matrix synthesis, mineralization.
↑ expression of growth factors; stimulation of bone cementum and periodontal ligament regeneration; induction of bone formation, osteoblast proliferation and differentiation
Aloe veraIn vitro (mesenchymal stem cells)
In vivo (rats)
[81]
Ellagic acid
Plants 12 02055 i016

Caffeic acid
Plants 12 02055 i017
- inhibition of iNOS, COX-2, NO, TNF-α, PGE2 and IL-6
- down-regulation of IL-1β-stimulated matrix metalloproteinase-13 and thrombospondin motifs 5
- up-regulation of collagen of type II and aggrecan
- suppression of NF-κB signaling

- ↓ chitinase-3-like protein-1, IL-1β, NF-κB, caspase-3; lipid peroxides, NO
- ↑ reduced glutathione
In vivo (mice)
In vitro (human chondrocytes)


In vivo (rats)
[82]





[83]
Ginsenoside
Plants 12 02055 i018
Modulatory compounds of bone regeneration pathways↑ calcium absorption at the intestinal level → thus prevents bone loss
↑ the level of trabecular calcium
↓ C-terminal telopeptide of type I collagen → ↓
resistance to tartrate acid phosphatase at the femoral level

Orchidaceae family

In vitro (mesenchymal stem cells)

[61,84]
Berberine
Plants 12 02055 i019
↓ bone loss by preventing decalcification and demineralization
inhibits osteoclastogenesis
suppresses the activity of the markers involved in the differentiation of acid phosphatase-resistant tartrate bone cells and cathepsin K
↓ the differentiation rate of osteoclasts
restore downregulation of osteogenesis-related genes expression;
↑ expression of osteogenesis-related genes such as OSX, COLⅠ, ALP, OCN and OPN
↑ total β-catenin and nuclear β-catenin; activation of the Wnt/β-catenin signaling pathway
Coptis species.
Berberis species.
Coptidis Rhizoma, Coptis chinensis, Coptis teeta.
In vitro (mesenchymal ctem cells)
In vitro (osteoblast and osteoclast)


In vitro (mesenchymal stem cells)
[85,86,87,88]




[87]
Apigenin
Plants 12 02055 i020
↑ the proliferation capacity of osteoblasts
inhibits decalcification and osteoclastogenesis
modulates intracellular signals → ↓bone loss induced by estrogen hormones
↓ the level of bone inflammation.

↑ mRNA levels of osteogenic genes BMP-2, Runx2 and COL1
downregulation of miR29a, miR17 and miR20a
Olea europaea.
Cassia occidentalis
In vivo (rats)
In vitro (osteoblasts)


In vitro (osteoblasts)
In vivo (rats)
[89,90,91,92]



Chlorogenic acid
Plants 12 02055 i021
↑ the level of favorable markers for bone formation
↑ the level of bone morphogenetic protein →↑ the activity of osteoblasts
↓ the level of pro-inflammatory factors
↑ the level of glutathione peroxidase →strong antioxidant effect
↑ the serum activity of alkaline phosphatase, osteoprotegerin
↓ the production of RANKL decreases
Prunus domestica L.In vivo (rats)
In vivo (rats)
[93,94]
Aesculetin
Plants 12 02055 i022
↑ expression of bone morphogenetic protein-2,
collagen type 1, osteoprotegerin;
ALP activation; transcription of Runt-related transcription factor 2; induction of: non-collagenous proteins of bone sialoprotein II, osteopontin, osteocalcin, and osteonectin, of annexin V and PHOSPHO 1.
↑ the production of thrombospondin-1 and tenascin C
-In vitro (osteoblasts)[95]


Acemannan
Plants 12 02055 i023


↑ mRNA expression of bone morphogenetic protein 2
↑ mineral deposition


Aloe vera

In vivo (volunteers)


[96]
Antihemorrhagic plant extract↑ osteoblastic activity and new bone formation; ↑ osteonectin and osteopontin expression

↓ inflammatory cell infiltration, vascular dilatation and hemorrhage
Glycyrrhiza glabra, Vitis vinifera, Alpinia officinarum Urtica dioica, Thymus vulgarisIn vivo (rats)[97]
Withaferin A
Plants 12 02055 i024
↑ expression of osteoblast-specific transcription factor and mineralizing genes, osteoblast survival,
↓ inflammatory cytokines.
Withania somniferaIn vitro (osteoblasts)
In vivo (mice, rats)
[98]
Ecdysterone
Plants 12 02055 i025
↑ gene expression of the BMP-2/Smad/Runx2/Osterix signaling pathway, stimulates MC3T3-E1 cell proliferation In vitro (osteoblasts)
In vivo (rats)
[99]



Echinacoside
Plants 12 02055 i026




↑ the uterine weight and serum E2 levels,
↓ body weight and hydroxyproline serum levels



Cistanche tubulosa



In vivo (rats)



[100]
Epigallocatechin gallate
Plants 12 02055 i027
activation of β-catenin of the Wnt signaling pathway
↑ expression of osteogenic genes, ALP activity, and mineralization in bone marrow-derived mesenchymal stem cells
Grean teaIn vitro (adipose-derived stem cells, dedifferentiated fat cells)
In vivo (mice, rats)
[101,102]
Essential oilsblocking nuclear factor kappa B, p38, and c-Jun N-terminal kinase signaling
↓ production of nitric oxide in RAW264.7 cells, inhibited EAhy926 cell proliferation
↑ serum C-telopeptide collagen type I and osteocalcin
↑ plasma calcium and vitamin D3, bone mineral-density
Prevention of inflammation and oxidative stress
Hypericum perforatum;
Cinnamomum burmanini;
Thymus vulgari;
Rosmarinus officinalis.
Populus alba;
In vitro (macrophages, fibroblasts, osteoblasts)
In vivo (rats, mice)
[48]
Forskolin
Plants 12 02055 i028
activation of cyclic adenosine monophosphate (c-AMP) signalling in stem cellsColeus forskohliiIn vitro (mesenchymal stem cells)[103]
Gallotannin
Plants 12 02055 i029
interaction with ALP
growth of Saos-2 cells
Mangifera indica L.In vitro (osteoblasts) [104]










Ursolic acid
Plants 12 02055 i030











↑ trabecular parameters (BV/TV, Tb.Th and conn.D)
↓ SMI
↑ALP activity, osteogenic genes (Runx2, BMP-2, type 1 Col1 and Wnt3a)
stimulates Wnt/β-catenin signalling
osteoblast differentiation (activation of mitochondrial respiration)










Psidium guajava









In vitro (osteoblasts)
In vivo (rats)










[105]
Malvidin
Plants 12 02055 i031
Cyanidin
Plants 12 02055 i032
Delphinidin
Plants 12 02055 i033
inhibition of MSC adipogenesis and downregulation of FABP4 and adiponectin genes.
↑ accumulation of calcium deposits
upregulation of osteocyte-specific gene BMP-2 and Runx-2 expression
BerriesIn vitro (mesenchymal stem cells)[106]
Rutin
Plants 12 02055 i034
activation of Wnt/b-Catenin Signaling
↑ activity of ALP, Runx2, osterix, osteocalcin, bone morphogenetic protein 2, Wnt3a, and b-catenin
Morinda citrifolia (Noni)In vitro (murine myoblast cell line, human periodontal ligament cells)
In vivo (rats)
[107,108,109]
Rhamnogalacturonan-I
Plants 12 02055 i035
↓ intracellular accumulation of galectin-3
up-regulation of collagen type I alpha 1 (COL-Iα1), osteocalcin, sialoprotein.
down-regulation of RANKL, TNFα, IL-6, and IL-1β
Solanum tuberosumIn vitro (neutrophils and macrophages; osteoblasts)
In vivo (rats)
[79,80]











Crocin
Plants 12 02055 i036
Crocetin
Plants 12 02055 i037












↑ ALP activity and ALP mRNA expression in MSCs











Crocus sativus L.










In vitro (mesenchymal stem cells)











[110]
Sinapic acid
Plants 12 02055 i038
activation of TGF-β1/BMP/Smads/Runx2 signaling pathways => osteoblast differentiationCynanchi atratiIn vitro (macrophags)
In vitro (mesenchymal stem cells)
In vivo (rats)
[111,112]
BetaEcdysone
Plants 12 02055 i039
↑ collagen deposition, ↑ levels of osteocalcin, ↑ expression of osteogenic genesTinospora cordifoliaIn vitro (osteoblasts, macrophages)
In vivo (rats)
[113]
Cucurbitacin B
Plants 12 02055 i040
↑ expression of ALP and OPN genes, mineralization
up-regulation of VEGFR2 and VEGFR-related signaling pathways (induction of angiogenesis)
Cucurbitaceae family plantsIn vitro (mesenchymal stem cells)
In vivo (rats)
[114]
Polysaccharideshematopoiesis protection:
↓ myeloid cells within tumor-infiltrating immune cells
Inhibition of hematopoietic cell expansion in the spleen
↑ HSPCs (hematopoietic stem and progenitor cells) and common lymphoid progenitors in the bone marrow
Polygonatum sibiricumIn vivo (mice)[115]
Ellagic acid
Plants 12 02055 i041
Ellagic acid and
Sennoside B
Plants 12 02055 i042

↑ number of osteoblasts and expression of OCN and OPG
↓ number of osteoclasts and the expression of RANKL














- repression of c-Jun expression at the mRNA level


In vivo (rats)















In vitro (human osteosarcoma cells)


[116]















[117]
Ellagic acid and hydroxyapatite↑ in the expression of FGF-2, VEGF and ALP
↑ IL-10, BMP-4 and OPN
↓ TNF-α and increasing the expression of
In vivo (rats)[118,119]
Melibiose
Plants 12 02055 i043
Methylophiopogonanone A
Plants 12 02055 i044
Tubuloside A
Plants 12 02055 i045
↑ expression of ALP, osteocalcin, osterin, osteoprotegerin, and autophagy marker proteins
activation of BMP2/Smad/Runx2 and Wnt/β-catenin signaling
Juglans regiaIn vitro (mesenchymal stem cells)[120]
RANK: receptor activator of nuclear factor kappa B; RANKL: receptor activator of nuclear factor kappa B ligand; BMP: bone morphogenetic protein; ALP: alkaline phosphatase; MSCs: Mesenchymal stem cells; APN: Adiponectin; COL-Iα1: collagen type I alpha 1; FABP4: fatty acid binding protein 4; RUNX2: runx-related transcription factor 2; ROS: reactive oxygen species; IL: interleukin; TGF-β: Transforming growth factor-beta; VEGF: vascular endothelial growth factor; JNK: c-jun-N-terminal kinase; E2: endogenous 17-β-estradiol; Wnt: the Wingless-type MMTV integration site family; PPARγ2: peroxisome proliferator-activated receptor γ2; SIRT 1: sirtuin 1; OSX: Osterix; OPN: osteopontin; SMAD: Small mother against decapentaplegic; iNOS: inducible nitric oxide synthase, COX-2: cyclooxygenase-2, NO: Nitric oxide, PGE2: prostaglandin E2, NF-κB: nuclear factor kappa B, OCN: osteocalcin protein, OPG: osteoprotegerin, OPN: osteopontin. Chemical structures were obtained from the site https://molview.org/ accessed on 17 May 2023.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hanga-Farcaș, A.; Miere, F.; Filip, G.A.; Clichici, S.; Fritea, L.; Vicaș, L.G.; Marian, E.; Pallag, A.; Jurca, T.; Filip, S.M.; et al. Phytochemical Compounds Involved in the Bone Regeneration Process and Their Innovative Administration: A Systematic Review. Plants 2023, 12, 2055. https://doi.org/10.3390/plants12102055

AMA Style

Hanga-Farcaș A, Miere F, Filip GA, Clichici S, Fritea L, Vicaș LG, Marian E, Pallag A, Jurca T, Filip SM, et al. Phytochemical Compounds Involved in the Bone Regeneration Process and Their Innovative Administration: A Systematic Review. Plants. 2023; 12(10):2055. https://doi.org/10.3390/plants12102055

Chicago/Turabian Style

Hanga-Farcaș, Alina, Florina Miere (Groza), Gabriela Adriana Filip, Simona Clichici, Luminita Fritea, Laura Grațiela Vicaș, Eleonora Marian, Annamaria Pallag, Tunde Jurca, Sanda Monica Filip, and et al. 2023. "Phytochemical Compounds Involved in the Bone Regeneration Process and Their Innovative Administration: A Systematic Review" Plants 12, no. 10: 2055. https://doi.org/10.3390/plants12102055

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

Article Metrics

Back to TopTop