Next Article in Journal
Preparation and Characterization of Patch Loaded with Clarithromycin Nanovesicles for Transdermal Drug Delivery
Next Article in Special Issue
Collagen Membranes Functionalized with 150 Cycles of Atomic Layer Deposited Titania Improve Osteopromotive Property in Critical-Size Defects Created on Rat Calvaria
Previous Article in Journal
Polysaccharide-Based Hydrogels and Their Application as Drug Delivery Systems in Cancer Treatment: A Review
Previous Article in Special Issue
An Experimental Anodized Titanium Surface for Transgingival Dental Implant Elements—Preliminary Report
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Localized Ionic Microenvironment in Bone Modelling/Remodelling: A Potential Guide for the Design of Biomaterials for Bone Tissue Engineering

1
Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
2
The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
3
School of Medicine and Dentistry & Menzies Health Institute Queensland, Griffith University (GU), Gold Coast, QLD 4222, Australia
*
Author to whom correspondence should be addressed.
J. Funct. Biomater. 2023, 14(2), 56; https://doi.org/10.3390/jfb14020056
Submission received: 3 November 2022 / Revised: 11 January 2023 / Accepted: 14 January 2023 / Published: 19 January 2023
(This article belongs to the Special Issue Biomaterials and Bioengineering in Dentistry)

Abstract

:
Bone is capable of adjusting size, shape, and quality to maintain its strength, toughness, and stiffness and to meet different needs of the body through continuous remodeling. The balance of bone homeostasis is orchestrated by interactions among different types of cells (mainly osteoblasts and osteoclasts), extracellular matrix, the surrounding biological milieus, and waste products from cell metabolisms. Inorganic ions liberated into the localized microenvironment during bone matrix degradation not only form apatite crystals as components or enter blood circulation to meet other bodily needs but also alter cellular activities as molecular modulators. The osteoinductive potential of inorganic motifs of bone has been gradually understood since the last century. Still, few have considered the naturally generated ionic microenvironment’s biological roles in bone remodeling. It is believed that a better understanding of the naturally balanced ionic microenvironment during bone remodeling can facilitate future biomaterial design for bone tissue engineering in terms of the modulatory roles of the ionic environment in the regenerative process.

1. Introduction

A localized microenvironment in bone remodeling milieus is generated and maintained when ions and biological molecules are released during the demineralization and degradation of bone matrix by protons and proteases secreted by osteoclasts, respectively, and bone formation by osteoblasts [1]. However, the localized microenvironment will be altered at implantation sites, with biomaterials interacting with extracellular fluid and cells. Considered vehicles for localized delivery of inorganic ions and ionic groups, inorganic biomaterials are no longer merely an inert scaffold but a reservoir for bioactive cues for modulating the bone remodeling process [2,3,4].
Inspired by the abundance of elements in the biological system and the effects of nutritional deficiency or overload, therapeutic applications of bioinorganic ions have been explored for many years. For example, the platinum drug cisplatin has been used for cancer treatment, the gold drugs myocrisin and auranofin for rheumatoid arthritis treatment, silver compounds in the pharmaceutical industry for their antimicrobial properties, and lanthanides and some transition metals as radiopharmaceuticals and diagnostic agents [5,6,7]. Meanwhile, the non-scientific and unregulated usage of inorganics can sometimes also be poisonous and lead to tragic disorders or diseases. For example, grey-colored skin is caused by unsafe nasal sprays due to the precipitation of silver salts, and copper deficiency results from over-supplemented zinc for prostate problems and acne [4,5]. In the field of regenerative medicine, the roles of elements in modulating cellular activities have gradually been unraveled, either as essential cofactors of enzymes and proteins or as regulatory molecules in ion channels or secondary signaling. Uncovered biological roles of ions provided possibilities to explore the applications of inorganic biomaterials in hard and soft tissue engineering by acting as vehicles to deliver ions and ionic groups locally.
Among all, calcium phosphates (CaPs) based on inorganic biomaterials are one of the most extensively studied types for bone grafting. They are composed of calcium ions and phosphate groups, which are omnipresent in the bloodstream or fixed in the bone mineral phase [8,9,10]. These synthetic bone substitutes can bind with natural bones by forming a solid biomaterial-bone interface, lacking osteoinductive and angiogenic properties [3,11,12,13,14]. Significant progress has been made in designing functional CaPs-based biomaterials with: (a) optimized geometry, roughness, and appropriate porosity for entrapping and concentrating growth factors or osteoprogenitor cells via proteins that could enhance cell adhesion, (b) incorporated growth factors or proteins that could modulate cellular activity, (c) doped trace elements that enhance osteogenesis in vitro [15,16]. However, the clinical performances of current CaP-based biomaterials are still unsatisfactory and incomparable to autologous bone grafts due to low bioactivities [8,10]. Nevertheless, the optimization of CaPs-based biomaterials significantly boosted the understanding of the modulatory effects of ions in the biological system [3,4]. Considering the abundance of ions in the bone environment and the current knowledge of their modulatory roles in maintaining the bone remodeling balance, it is expected that a deeper understanding of ions in the bone environment would provide new insights to guide the future design of inorganic biomaterials for bone tissue engineering [3,17]. In this review, we focus on inorganic components in the bone environment, helping to provide new insights on how it might be profound to guide the future design of inorganic biomaterials for bone tissue engineering.

2. Bone Mineral Phase and Localized Ionic Microenvironment

Bone homeostasis is maintained in a series of highly complicated events orchestrated by: (a) interactions among different types of cells, mainly mesenchymal stem cells (MSCs), osteoprogenitor cells, osteoblasts, osteoclasts, and osteocytes, and (b) interactions of cells with extracellular matrix in a localized microenvironment, and (c) interactions of cells with components in surrounding biological milieus, such as organics (amino acids, enzymes, hormones, fatty acids, neurotransmitters, sugars, vitamins, etc.), inorganics (inorganic ions or groups, such as calcium, phosphate, potassium, sodium, carbonate, etc.), as well as waste products from cell metabolism (Figure 1) [9]. Naturally, the localized ionic microenvironment is maintained by the balance between bone-forming cells, osteoblasts, and bone-resorbing cells, osteoclasts, during the bone remodeling process [18] (Figure 2). Specifically, osteoclasts firmly attaching to the bone surface could achieve a pH fall to a limit value of pH 3.0 or less for dissolving the bone mineral and favor collagen degradation by secreting lysosomal proteinases [19]. Organics in this microenvironment have been extensively studied since the last century, especially cell-secreted growth factors that play roles in bone formation, such as TGF-β (transforming growth factor-beta), FGF (fibroblast growth factor), BMP (bone morphogenetic proteins), IGF-I (insulin-like growth factors I), etc. [20]. Inorganics in this microenvironment have also been extensively studied because they are essential for the bone mineral formation, and quality of the mineralized tissue, either liberated from bone or circulating in body fluid. Moreover, ions from the localized microenvironment are now considered to consist of crystal components and molecular modulators in many biological processes in bone remodeling, i.e., bone formation and resorption [3,9,21,22]. The list of inorganic ions and ionic groups that affect bone metabolism and homeostasis as signaling molecules has dramatically increased in the past decades. More previously less-studied elements in the periodic table have been surprisingly found to play a role in the etiology and pathogenesis of some bone diseases or the modulation of cellular activities, especially metallic elements, because they are prone to lose electrons to form positively charged ions and tend to dissolve in biological fluids or be attracted by negatively charged biological molecules, proteins, or DNAs, to form active metal complexes [3,4,23].
Bone mineral, known as biological apatite, is incorporated in collagen fibrils, arranged with a c-axis parallel to the direction of fibrils, with lengths of 30–50 nm, widths of 15–30 nm, and thicknesses of 2–10 nm [24]. Biological apatite has been modeled as hexagonal carbonated hydroxyapatite based on X-ray diffraction (XRD) results, with the lattice parameters of a = b = 9.432 Å, c = 6.881 Å, and γ = 120°. Hydroxyl ions (OH), parallel to the c-axis, are positioned on the screw axes at every one-half of the unit cell, pointing in opposite directions to neighboring OHs. Tetrahedral phosphate ions (PO43−), immobilized by calcium ions (Ca2+) interspersed among them, as well as marginal calcium ions (Ca2+), shared with neighbor unit cell. Notably, steric interference between adjacent OHs in hexagonal hydroxyapatite unit cells could be overcome by vacancy or replacement of an OH by impurity ions, the most likely event in organisms, or by conversion of hexagonal to monoclinic space group at high temperature, rearranging adjacent OHs to a uniform direction [25,26].
The accumulation of most inorganics in the body can be attributed to the formation of apatite crystals in bone, with distinguished content and composition among species and individuals, resulting from differed preferences on elements in different species, variations in diet, and relative abundance in the environment [27,28]. During bone resorption, ions and ionic groups will be liberated from CaP based network into the local microenvironment in acidic conditions, participating in local bone remodeling or being carried away by physiological fluid. Therefore, ions and ionic groups entering the localized microenvironment are determined by the composition of bone minerals and vice versa. Specifically, the content and level of ions and ionic groups in the localized biological milieu affect the formation of the bone mineral through ionic substitution and, consequently, the properties of the final crystalline product in the mineral phase [29,30,31,32]. For example, OH (minor site) or PO43− (primary site) sites could be replaced by CO32−, forming type A and B carbonated hydroxyapatite, respectively. PO43− site could also be replaced by hydrolyzed phosphate (HPO42−) structure in mature bone, while OH could be substituted with florin (F), chlorin (Cl) ion, or orthosilicic acid (SiO44−) structure [29,30,32]. Moreover, ionic substitutions also happen where Ca2+ is replaced by other metals, such as sodium (Na+), potassium (K+), magnesium (Mg2+), zinc (Zn2+), manganese (Mn2+), cobalt (Co2+), strontium (Sr2+), iron (Fe2+), copper (Cu2+). Ionic exchange in biological apatite alters crystalline structures, resulting in modified crystal size, growth rate, and properties. Compared with stoichiometric or geological apatite crystals, biological apatite crystals have smaller crystallite sizes, less ordered crystal structure, lower crystallinity, and higher solubility (Figure 3) [3,33]. The influences of different ions or ionic groups on biological apatite crystals are balanced by each other. For example, substitutions of PO43− by CO32− and of Ca2+ by Zn2+ or Mg2+ inhibit crystal growth, increase crystal disorder and solubility, and lower the crystallinity [29,30,34]. Replacements of Ca2+ by Al3+, La2+, or Fe2+ accelerate crystal growth, and replacement of OH by F on the lattice reduces the solubility [29,30,34]. Additionally, replacing OH with SiO44− causes a contraction on the a-axis and an expansion on the c-axis of the crystal lattice [35]; replacing Ca2+ with Sr2+ causes an expansion on both the a- and c-axes [4].
During bone trauma, such as a fracture, bone healing starts with the invasion of blood into the traumatic space. A microenvironment is formed along with blood clots and calluses, where cells interact with components in the extracellular matrix and extracellular fluid. However, the localized microenvironment will be altered at the implantation site with the involvement of inorganic biomaterials due to extensive interactions between biomaterials and the microenvironment [16]. The contribution of inorganic biomaterial at the implantation site to the healing process can never be underestimated because many biomaterial intrinsic features, including parameters (composition, structure, topography), and properties (crystallinity, dissolution profile, surface charge), can make a difference in the localized microenvironment and cellular interactions, as well as cellular activities, and consequently the bone formation process (Figure 4) [16]. Therefore, understanding the influence of biomaterials on components in the localized ionic microenvironment shall guide the design of future inorganic biomaterials for bone grafting. Active roles of ions as molecular modulators upon many cellular activities during bone remodeling provide the material with more possibilities other than structural support and protein/cell entrapping.

3. Active Osteoinductivity of Inorganic Biomaterials and Enriched Localized Microenvironment

There are a variety of commercial substitute materials for bone and tooth repair/replacement, including metals, polymers, corals, processed human or animal bones, synthetic CaP materials such as ceramics or cement, and hybrid composites [16]. CaPs are one of the most extensively studied inorganic materials for bone grafting due to the omnipresent presence of calcium ions and phosphate groups in the bloodstream or bone. They are excellent in biocompatibility, osteoconductivity, and osteointegration but are brittle and unsuitable for load-bearing [36]. The first attempt to repair surgically created defects in rabbits with artificial CaP material (TCP) was in 1920 [37]. In 1975, β-TCP was applied for the first time in a surgically created periodontal defect in dogs and as an adjunct to apical closure in pulpless permanent teeth in humans [38,39,40]. The first attempt to replace tooth roots with synthetic dense HAp cylinders was reported in 1979 [41]. However, the popularity of CaPs as substitute xenografts or allografts did not start until the late 1990s, which were strictly controlled due to the consequent appearance of diseases after implantation, such as acquired immunodeficiency syndrome (AIDS), and bovine spongiform encephalopathy [2,23,42,43].
Bioactive glasses with a modified SiO2 network, developed by Larry L. Hench in the late 1960s, are another group of extensively investigated inorganic biomaterial as an implant for bone defects over the years and achieved great success in the clinical field [44]. Many commercially available synthetic inorganic biomaterials are primarily obtained via wet chemistry, starting from a mixture of ionic solutions or solid-state conversion with heat treatment. In addition to the synthetic method via wet chemistry, inorganic biomaterials can also be obtained from nature. For example, CaP-based biomaterials could be obtained from chemically similar marine coral via hydrothermal conversion of the calcium carbonate skeleton of marine coral to hydroxyapatite. Coral-derived hydroxyapatite has been used as a bone graft since the 1980s for good biocompatibility and structural support. Still, it was limited in clinical practice due to inherent weak mechanical strength and low degradability [45,46]. Moreover, the composition of marine coral-derived hydroxyapatite also differs from that in the natural bone mineral phase [47]. Elements in the bone mineral phase are constantly fixed and liberated during bone remodeling. Ions involved in amorphous calcium phosphate formation at the early stage of nucleation, phase transition during crystallization, and extensive ionic substitutions along the mineralization process during bone formation, are from the local microenvironment at the implantation site, i.e., bloodstream and implanted material. These ions are essential to the body and are considered bioactive ions not only because they form the bone mineral phase through crystallization, but also because they participate in modulating multiple cellular activities in bone metabolism, such as the proliferation and differentiation of osteoblasts and osteoclasts, as well as the responses of immune cells [48,49,50,51,52,53,54]. Therefore, it is speculated that biomaterials obtained from bone are most likely to achieve the maximum retainment of bioactive trace elements by retaining bioactive ions originally present in bone. There are several bone mineral products on the market, mainly in dentistry, such as Cerabone (AAP Biomaterials GmbH, Berlin, Germany) and Bio-Oss (Geistlich Pharma AG, Wolhusen, Switzerland) [11]. Cerabone®, a bone mineral product of bovine origin manufactured by a proprietary 1200 °C production process, is mainly used to support the successful placement of dental implants. And Bio-Oss®, primarily used in dental surgery, is obtained by removing organic substances with a stepwise annealing process up to 300 °C, followed by a strong alkali treatment [43,55]. Removal of viruses, bacteria, proteins, and other organic substances via sintering not only leaves a three-dimensional porous network, facilitating protein adsorption and cell adhesion but also increases the crystallinity of mineral crystals with reduced solubility and improved mechanical strength and biological stability. However, the degradation of this product type is considerably slow, with visible remnants in the 30-month post-implantation [56].
After implantation, graft materials are expected to allow bone-like apatite to deposit on the surface-mediated by cells, and consequently bond to surrounding living bone, obtaining extra stabilization and fixation at the implant region [57,58]. Currently, the most widely adopted approach to predict bone-bonding ability (i.e., osteoconductivity) is to test the ability to deposit bone-like apatite on the surface of a material by immersing in simulated body fluid (SBF), an ionic solution with nearly equal ion concentrations to those of human blood plasma [57]. Graft materials are also expected to be excellent in osteointegration, as biodegradation and biosorption favor vascular and bony ingrowth and cellular waste removal. In addition to osteoconductivity and osteointegration, osteoinductivity, the ability to induce new bone growth, is another essential property of graft material. The osteoinductive property of a biomaterial is usually demonstrated by de novo bone formation in the absence of osteogenic factors and non-osseous sites after implantation in vivo.
In general, osteoinductivity of inorganic biomaterials can be obtained from (a) material design with proper geometry, roughness, and porosity that facilitates bone growth by entrapping and concentrating growth factors or osteoprogenitor cells, (b) incorporation with growth factors, bioactive proteins or trace elements that would induce bone growth [3,59]. If osteoinductivity obtained through the optimization of parameters and biophysical properties of an inorganic biomaterial via entrapped or concentrated growth factors or osteoprogenitor cells is considered “passive osteoinductivity”, osteoinductivity obtained by incorporating osteogenic proteins or bioactive inorganics should be considered as “intrinsic osteoinductivity”, because molecules, such as growth factors, and bioactive trace elements, liberated from materials with active osteoinductivity participate in new bone formation proactively via modulating cellular activities.
Osteoinductivity of demineralized bone matrix in different animals was reported in 1965, and osteogenic factors originally present in the matrix, specifically bone morphogenetic proteins (BMPs), were demonstrated later [60,61]. Inorganic biomaterials incorporated with BMPs, sourced from extraction or recombinant procedures, have been investigated extensively for many years due to their excellent osteoinductivity [62,63,64,65,66]. Other biological osteogenic/angiogenic factors have also been extensively studied over the years, such as TGF-β, FGF, VEGF (vascular endothelial growth factors), parathyroid hormone, and PRP (platelet-rich plasma) [3,15,23,67,68,69]. Numerous combinations of growth factors and types of inorganic biomaterials have been explored for intrinsic osteoinductivity and angiogenesis, as summarized in many reviews [10,16,68,70]. However, inorganic biomaterials incorporated with growth factors are mainly limited in clinical application, with increasingly raised safety concerns regarding the off-label usages of growth factors and their high costs [4].
With intrinsic properties of inorganic biomaterials to release ions, osteoinductive inorganic biomaterials can also be achieved via increased ion concentration in the localized microenvironment. In general, the liberation of ions from biomaterials is believed to enrich the ionic microenvironment, alter ion concentrations and local pH and get involved in bone formation by increasing the supersaturation of ions toward the deposition of hydroxyapatite or as molecular modulators to affect cell signaling and activities [3,4,16,54]. The mineralization on the surface of inorganic biomaterial after implantation, as well as the process of bone formation, is affected by cytotoxicity and osteoconductivity of the material and the impact of it upon cellular activities by releasing ions and ionic groups into the local microenvironment, i.e., the biological milieus. A schematic illustration is shown in Figure 4 to explain the dissolution and precipitation process near the surface of biomaterial in vivo in the ionic microenvironment created by physiological fluid and enriched by dissolved biomaterial. Specifically, ions and ionic groups are liberated from biomaterial either through solubility-determined dissolution in the physiological environment or cell-mediated dissolution in the acidic environment created by macrophages or osteoclasts, resulting in localized supersaturation of inorganics in the microenvironment, further leading to the precipitation of calcium-deficient HAp [4,8,16]. Inspired by the observed integration of biomaterials with the host bony tissues via the deposition of HAp, simulated body fluid (SBF) was developed to predict in vivo bone-bonding activity near the surface of the implanted biomaterial [8,57]. The standardized SBF solution contains a similar ionic profile as the blood and showed a good correlation between the in vivo bioactivity of bioactive glass and apatite-forming ability in the early years [57,71]. Some concerns were proposed in recent years regarding the validity of the SBF immersion test by Bohner et al. and Pan et al. [72,73]. For example, the interference of proteins on apatite formation and the control of carbonate content is not considered [72]. In addition, the roles of ions and ionic groups in the localized biological milieu are also underestimated because they are never merely components in forming mineral crystals, aggregating freely to reach a relatively stable state with lower energy, but also modulators of various cellular activities, such as the proliferation and differentiation of osteoblasts/osteoclasts and getting involved in the crystal formation process on the surface of biomaterials [4,8,16].

4. Summary of Ions and Ionic Groups in the Maintenance of Bone Homeostasis

Inorganic ions are not only nutrients in the body but also have the potential as components in diagnostic or therapeutic agents to study or treat various diseases and metabolic disorders, explaining why they have great potential to affect bone regeneration to a similar extent as recombinant growth factors but free from safety issues [4,23]. For example, calcium and phosphate are essential in bone and many other biological processes. Sufficient calcium intake from food or supplementation contributes to maintaining calcium homeostasis in the body, promotes mineralization during growth, and reduces bone loss in the elderly. In contrast, prolonged calcium deficiency may lead to rickets, osteomalacia, and osteoporosis [74]. Similarly, long-term inorganic phosphorus deficiency causes hypophosphatemia, impaired bone mineralization, dysfunction in the blood, muscle, and central nervous system, and the cardio and respiratory system [75]. In skeletal bone, the local availability of both ions is one of the determinants for extracellular matrix mineralization rate, the last step of the bone formation process, and regulatory molecules for multiple cellular activities. However, the bioactivities of other ions were significant and cannot be overlooked. The biological influences of ions at both physiological and cellular levels have been summarized in Table 1 and Figure 5. Notably, there are some limitations in these studies and should be considered in future studies: (a) the ionic profile in the cell culture media upon the addition of ions was overlooked in most studies; (b) the discrepancy/consistency between in vitro and in vivo studies should be emphasized; (c) the justification of whether phenomenon observed in the investigation is caused by the ion of interest.

4.1. Extracellular Calcium-Ca2+

Extracellular Ca2+ has been shown to correlate with multiple cellular activities of MSCs (growth, osteogenic differentiation, and mineralization), osteoblasts (survival, proliferation, and differentiation), and osteoclasts (survival and bone resorption activity) via a variety of intracellular signaling pathways in vitro [4,54,164,165]. In MSCs, the optimized concentration of Ca2+ is 1.8 mM, the same concentration supplied in culture media to maintain cell growth [76]. The microenvironment of Ca2+ at a concentration <1.8 mM significantly impeded cell growth and osteogenic differentiation [76]. Higher Ca2+ concentration, on the other hand, showed no additional promotive effect on cell growth but affected the extent of cell mineralization in a dose-dependent manner [76]. The fluctuation of extracellular Ca2+ concentration is most likely to be sensed by the functional calcium-sensing receptor (CaSR) on the cell membrane, which is a member of the G-protein-coupled receptor (GPCR) superfamily [79,164,165]. CaSR is believed to be critical in maintaining the homeostasis of extracellular Ca2+ concentration and modulating cell metabolism in many cells, such as parathyroid gland cells, kidney cells, bone cells, endothelial cells, and stem cells [164,166]. In osteoblast cells, Ca2+ has been suggested to promote osteoblast proliferation and survival (2–4 mM), as well as differentiation (~5 mM), with elevated expression of several osteogenic markers such as type I collagen (Col-I), bone morphogenetic proteins (BMP), osteocalcin (OCN), etc., most likely via CaSR-mediated signaling pathways [77,78,164,165,167,168,169,170,171]. The proliferation of osteoblast is associated with the activation of extracellular signal-regulated kinase-1 and -2 (ERK-1 and ERK-2) signaling pathways from the mitogen-activated protein kinase (MAPK) superfamily through dual phosphorylation of critical threonine and tyrosine residues [77,78]. The inhibition of osteoblast apoptosis is attributed to the activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB) pro-survival pathway [77,78]. Other intracellular signaling pathways, such as phospholipase C (PLC) and protein kinase C (PKC), Jun-terminal kinase (JNK), and cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA), are also activated through CaSR in high Ca2+ environment to affect cell survival [69,172,173]. In addition, the expression of several secondary messengers can also be induced by CaSR signaling pathways, mediating extracellular Ca2+ level and controlling osteoblast cell fate, such as insulin-like growth factor (IGF)-II (required for the subsequent cell proliferation), or prostaglandin E2 (PGE2) produced by cyclooxygenase-2 (COX-2) (associated with alkaline phosphatase activity, and the expression of osteocalcin) [173,174,175]. Moreover, Ca2+ has also been shown to regulate cell morphology via cell-cell or cell-matrix interaction, enhancing the expression of angiopoietin-1 (Ang1) and angiogenesis [166,167,176]. In osteoclasts, internalized through CaSR, Ca2+ has been shown to sequentially activate the PLC signaling pathway, followed by PLC-dependent translocation of nuclear factor-κB (NF-κB) from the cytoplasm to the nucleus of mature osteoclasts and consequently induce cell apoptosis to inhibit bone resorption [9,79,80].

4.2. Inorganic Orthophosphate—Pi

The homeostasis of phosphate in the body is maintained by the cooperation of the gut, bones, and kidneys and balanced by parathyroid hormones, but limited knowledge from the entry beyond regarding the sensing mechanism and consequent proposal for appropriate regulation cascade [75,177]. The optimized concentration of Pi for MSC growth in vitro was proposed to be 0.09 mM; either higher or lower concentration caused impeded growth but showed little effect on cell differentiation or mineralization [76]. In cultured osteoblasts, Pi is found to be involved in modulating cell proliferation and DNA synthesis in a dose-dependent manner at a concentration from 2 to 4 mM, in part via the IGF-1 signaling pathway [178,179]. Pi is also found to regulate osteoblast differentiation and bone mineralization. For example, it induces the production of osteopontin (OPN), a molecule involved in the regulation of bone mineralization, through the activation of both ERK1/2- and PKC- dependent signaling pathways, as well as alkaline phosphatase (ALP) activity in vitro [180]. Pi stimulates the expression of stanniocalcin 1 (STC1, a regulator for the accumulation of transcription factor), pituitary-specific positive transcription factor 1 (Pit1), and consequently increases Pi uptake and mineralization both in vitro and in vivo [82]. In addition, it stimulates the production of matrix Gla protein (MGP, one of the key regulators in extracellular mineralization) with the involvement of Ca2+ via ERK1/2- dependent signaling pathways and upregulates the expression of Fos-related antigens 1 and 2 (Fra-1/2) of activator protein-1 (AP-1) family in vitro [181,182,183]. The Pi-promoted osteoblastic differentiation and mineralization provided theoretical support for the localized delivery of Pi from implant materials to promote mineralization [81,82]. However, a high Pi microenvironment resulting from bone resorption or material dissolution may cause significant osteoblast apoptosis through the induction of a transition on mitochondrial membrane permeability, in accordance with observed osteoblast cell apoptosis at bone resorption sites [50,83]. In osteoclasts, osteoclastic bone resorption is well known to be stimulated by low Pi concentration but inhibited with the increase of Pi level [84,184]. The inhibitory effect of Pi on the bone resorptive activity at higher concentrations can be partially attributed to the direct induction of osteoclast cell apoptosis and the inhibition of receptor activator of nuclear factor kappa-Β ligand (RANKL)-induced JNK and Akt signaling pathways [84,85].

4.3. Other Bioactive Inorganic Ions

Boron (B), an essential micronutrient, is considered to play an important role in the maintenance of bone and osteogenesis. Several in vivo studies have shown that B ion deficiency could result in reduced osteogenesis, and B ion deprivation would inhibit bone formation, resulting in reduced bone volume and mechanical strength. Beneficial effects on bone micro-architecture and strength could be observed with the nutritional intake of B [87,88,89,90]. In addition to the effects of dietary B from previous in vivo studies, beneficial effects of B ion are also found at the cellular level in BMSCs and osteoblasts: B ion is capable of increasing osteogenic marker gene (ALP, OCN, and Col-I) expression and inducing early matrix mineralization in MSCs, and regulating osteogenic marker expression (runt-related transcription factor 2 (Runx2), and bone sialoprotein (BSP) at mRNA level, BMP-4, -6 and -7 at protein level) in osteoblasts [49,91,92].
Copper (Cu) is an essential trace element required for the function of several important enzymes in the body, and it is necessary to maintain bone quality and strength [185]. Cu2+ deficiency causes abnormal bone formation with impaired quality as a co-factor of an enzyme, lysyl oxidase [93]. It prevents crosslinking between structural proteins, collagen, and elastin; At the same time, excess Cu levels may generate free radicals, inducing lipid peroxidation and affecting bone metabolism, and may lead to severe neurological issues or liver diseases [93,94,95]. Nevertheless, being discovered as an essential element with angiogenic and innate antibacterial properties, the applications of Cu2+ have attracted much attention in biomaterial fabrication [4,96,97,98]. Rapid and enhanced vascularization and increased extracellular matrix formation are achieved with several Cu-doped biomaterials, bringing novel insights to the traditional concept of accelerating bone formation by filling pores instead of ingrowth from periphery regions [99].
Gallium (Ga) is not an essential element in the body but positively affects bone formation with profound anti-resorptive activity [100]. Ga ion is found to inhibit osteoclast differentiation and osteoclastic resorptive activity in a dose-dependent manner by blocking the transient receptor potential cation channel subfamily V member 5 (TRPV5) Ca2+ channel (essential for osteoclast bone resorption); Improved mineralization and elevated mechanical properties results from inhibited expression of nuclear factor of activated T cells, cytoplasmic 1 gene (NFATc1, a regulator in osteoclast differentiation) [53,101,102]. However, the bioavailability of Ga remains a challenge because Ga salts are prone to form hydroxides and are potentially harmful upon consumption [100].
Magnesium (Mg), the second most abundant intracellular cation, stabilizes DNA and RNA structures and cell membranes and plays an essential role in maintaining the function of many enzymes as co-factors [74,186]. In skeletal bone, Mg deficiency contributes to impaired bone growth, disrupted mineral metabolism, decreased osteoblast, increased osteoclast cell number, and osteoporosis in young animals, with promoted inflammation [103,104,105,106,107]. Mg2+ is found to enhance the expression of the osteogenesis-related genes, production of extracellular matrix, and deposition of apatite crystal in undifferentiated MSCs and osteoblastic MSCs in vitro, possibly through the upregulation of hypoxia-inducible factor (HIF) and peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α), respectively [108,109,110]. In vivo studies also showed enhanced bone regeneration with overexpressed osteogenic markers, OCN, Runx2, and IGF-I, around the implant in vivo [108,109,110]. Nevertheless, Mg2+ (up to 5 mM) competes with Ca2+ as an antagonist and forms an insoluble salt with pyrophosphate, causing mineralization defect and cell dysfunction [106,111,112].
Iron (Fe) is an essential element for humans. In skeletal bone, Fe contributes to the homeostasis of bone, with evidence showing that: Fe deficiency causes an overall loss in bone mass and density, with impaired biomechanical strength [113]; Fe overload is associated with disrupted differentiation and maturation of osteoblasts through the production of reactive oxygen species (ROS) [116]. Increased oxidative stress on cells elevates cytokine (tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6)) levels, leading to bone resorption and altered bone microarchitecture, and consequent bone loss and reduced biomechanical strength [114,115,116].
Manganese (Mn), an essential cofactor for many enzymes, is required in many biological processes. In skeletal bone, Mn deficiency causes abnormal bone growth (such as stunted bone growth and osteoporosis), while Mn overload leads to impaired bone development and neurotoxicity [21,117,118]. Incorporated in the inorganic biomaterial, Mn promotes the proliferation, adhesion, and spreading of osteoblasts, upregulates osteogenic-related gene expression (ALP, BMP), and accelerates collagen deposition [119,120,121,122]. Meanwhile, localized administration of Mn2+ exhibits an insulin-like effect, promoting angiogenesis and bone healing in vivo [123].
Selenium (Se) is an essential trace element in humans. The level of Se is correlated with bone metabolism and maintenance (Kashin-Beck disease), as well as well-being and protection against aging-related diseases [74,130,131]. Se deficiency leads to impaired bone and cartilage metabolism, osteopenia, and fracture susceptibility in several studies, both in vitro and in vivo, and even contributes to the progress of osteoporosis [124,125,126,127]. In contrast, Se overload is harmful to bone due to decreased mineral content, altered bone structure, and reduced biomechanical strength [128,129]. Both sides of the influence of Se on bone health are indications of the possible modulatory role of Se in the maintenance of skeleton bone. Se is likely involved in cellular responses in bone development by regulating microRNA in the formation of selenoprotein [187]. However, the roles of proteins and the influences of Se in bone metabolism at the cellular level remain unclear.
Silicon (Si), mainly found in the skeleton, is essential in bone metabolism [74,188,189]. In addition to the evidence of positive effects of dietary Si supplementation on bone health, promotive effects of Si-containing biomaterials in bone regeneration have also been extensively investigated [134,188,190,191,192]. Among all Si-containing biomaterials, bioactive glass is the most extensively studied. Bioglass 45S5 (BG), composed of SiO2, CaO, Na2O, and P2O5, was developed in the late 1960s [44]. Bioactive glasses are known for their excellence in bone bonding by forming an apatite layer on the surface and their capability to stimulate and promote the growth, proliferation, and differentiation of osteoblasts [132,133,134]. Soluble Si ions, in the form of orthosilicic acid, are found to stimulate osteogenic differentiation and enhance osteogenesis both in vitro and in vivo, possibly with the involvement of Wnt and Sonic Hedgehog (Shh) signaling pathways and the upregulation of miR-146a to antagonize the activation of NF-κB signaling pathway [135,136,137,138,139,140]. Si is also found to inhibit osteoclast phenotypic gene expression, osteoclast formation, and recruitment, as well as bone resorption in vitro, via reduced expression of receptor activator of nuclear factor-κB (RANK)/RANKL/osteoprotegerin (OPG) gene in osteoclast precursors or osteoclasts without the involvement of osteoblasts/stromal cells [51]. In addition, the inhibition effect of Si ions on osteoblast-induced osteoclastogenesis on murine macrophage cell line (RAW 264.7 cells) is also demonstrated in a co-culture system with human osteoblastic-like cell line (SaOS-2), resulting from increased secretion of OPG in osteoblastic-like cells and increased ratio of OPG/RANKL [138].
Strontium (Sr), mostly stored in skeleton bone, can exert many effects on bone metabolism at cellular and tissue levels in vitro and in vivo [145,193]. Strontium ranelate (SrRan), an organic salt of Sr, has been used as an anti-osteoporotic drug to treat osteoporosis for many years by shifting the balance between bone formation and resorption towards the former, although the mechanism remains unclear [142,146,194,195]. It is believed that SrRan enhances pre-osteoblast cell replication and collagen synthesis promotes osteoblast proliferation and differentiation and reduces bone resorption by reducing differentiation of osteoclasts and increasing osteoclast apoptosis, partly via the activation of CaSR due to the chemical similarity between Sr and Ca [141,142,145,146,147]. Sr2+ can activate the Wnt/β-catenin pathway or Ras/MAPK signaling pathway to upregulate the expression of osteogenic differentiation markers in cultured MSCs (such as ALP, Col-1, Runx2, OCN, and COX2), facilitate calcium deposition and nodule formation, and promote in vivo bone formation [48,143,144]. In osteoblasts, SrRan (1–5 mM) promotes cell survival and proliferation, depending on the activation of Akt- and ERK1/2- dependent signaling pathway via CaSR, or acts independently to modulate osteoblast viability and replication [78,141,142]. SrRan (0.1–1 mM) also induces differentiation in osteoblasts with observed overexpression of ALP, bone sialoprotein (BSP), OCN, and Runx2 [142,146]. Additionally, Sr2+ (20 and 100 μg/mL) was found to disturb mineralization in rodent MSCs [196]. In osteoblast-induced osteoclastogenesis, SrRan affects the balance between OPG and RANKL genes, further suppressing the NF-κB signaling in vitro and in vivo [148,149]. The direct impact of SrRan on osteoclasts involves the activation of NF-κB translocation and consequent mature cell apoptosis via the activation of the PKCβII signaling pathway in a dose-dependent manner; The inhibition of osteoclastic differentiation and resorptive activity is achieved by the reduction of carbonic anhydrase II (key enzyme for bone resorption) and vitronectin receptor (involved in the motility of osteoclast and maintenance of the sealing zone) expression [142,150,151].
Zinc [65] is an essential nutrient for the catalytic activity of over 200 enzymes in numerous biological processes, such as immune response, wound healing, and DNA and protein synthesis [197]. In skeletal bone, Zn is the most abundant trace metal and an essential cofactor for some bone metabolism-related enzymes, such as ALP (provides a phosphate source for bone mineralization) and collagenase and matrix metalloproteinases (essential in bone resorption and remodeling), indicating its role in maintaining bone mass, health and bone turnover rate [154,155]. In cultured hBMSCs, Zn2+ released from the implant material has been shown to promote cell viability, osteoblastic marker gene expression (Col-1, OCN, ALP, and BSP), matrix maturation, calcium deposition, and nodule formation [156]. Zn2+ is also believed to participate in bone metabolism as a signaling molecule modulating osteoblast and osteoclast cellular activities in vitro and in vivo [156,157,158]. In cultured osteoblasts, Zn2+ stimulates cell proliferation, differentiation, and mineralization by stimulating gene expression of various proteins associated with osteoblastic differentiation, such as type I collagen, ALP, OCN, OPN, and Runx2, and production of growth factors, such as IGF-1 or estrogen, related to enhanced cell proliferation [159,160]. In osteoclasts, Zn2+ acts as a potent inhibitor of resorptive activities [162]. The mechanisms of Zn2+ in promoting bone formation and suppressing bone resorption are achieved via the inhibition of the activation of TNFα driven NF-κB pathway [163].
In addition, the influence of some elements remains controversial due to conflicting results obtained in different studies, such as Fluorine (F), Lithium (Li), and Titanium (Ti). Some are being investigated due to observed positive effects in some therapeutical applications in bone diseases or biomaterial fabrication, such as Germanium (Ge), Niobium (Nb), and Vanadium (V) [74,185]. Some dose-related toxic metals in the body, released from inorganic biomaterials or dietary intake, have influenced bone resorption and formation through the modulations of bone cell activities. For example, Cobalt (Co) and Chromium (Cr), the major components of prosthetic implant materials for hip and knee joint replacements, are revealed to affect bone health with dissolved Co2+ and Cr3+ into the peri-implant bone and cause progressive local osteolysis [22,198,199]. Cytotoxicity of Co2+ and Cr3+ are well-established in osteoblast-like cells with altered morphology, decreased cell number, proliferation, and cellular activities with decreased release of OCN and collagen type I, reduced ALP activity and calcium deposition, possibly due to altered redox state [22,198,200,201]. Growth factors/cytokines, such as TGF-β1, TNF-α, IL-1β, and IL-6, secreted from osteoblasts under the stimulation of Co and Cr ions, lead to inflammation and further induce the maturation and differentiation of osteoclasts [199,200]. Nevertheless, the role of Co2+ in promoting vascularization in bone tissue is still worth pursuing because vascularization is also a critical component in bone regeneration [202,203,204,205].

5. Conclusions and Future Perspectives

A localized ionic environment is generated during bone remodeling, where the bone matrix containing organic molecules (growth factors, enzymes, etc.) and inorganic ions and ionic groups (Ca2+, PO43−, Mg2+, etc.) is degraded. There is a wealth of evidence revealing the osteoinductive potentials of many individual ions, but few consider the effect of the ionic microenvironment. The composition and interactions among components in the localized ionic environment during bone remodeling seemed challenging to investigate. Still, we believe the mystery will be unveiled with more emerging state-of-the-art techniques and a deeper understanding of related fields. Additionally, more fundamental research is needed to address the effective species and dosage during biomaterial fabrication in the future. Nevertheless, the beneficial effects of ions in bone tissue engineering will shed light on the design of future inorganic biomaterials for bone regeneration. An inorganic biomaterial that provides a balanced combination of inorganic ions in a controlled and sustained way will potentially generate a desired ionic environment to regulate bone cell functions, resulting in optimal tissue regeneration.

Author Contributions

Conceptualization and supervision, Y.X., Z.D., R.C. and Y.M.; original draft preparation, Y.M.; review and editing, Z.D., L.X., W.G., R.C. and Y.X.; funding acquisition, Y.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research is supported by the Joint Research Centre Fund from the Department of Environment and Science (2019–2023), Queensland.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Itzstein, C.; Coxon, F.P.; Rogers, M.J. The regulation of osteoclast function and bone resorption by small GTPases. Small GTPases 2011, 2, 117–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Habraken, W.; Habibovic, P.; Epple, M.; Bohner, M. Calcium phosphates in biomedical applications: Materials for the future? Mater. Today 2016, 19, 69–87. [Google Scholar] [CrossRef]
  3. Wang, W.; Yeung, K.W.K. Bone grafts and biomaterials substitutes for bone defect repair: A review. Bioact. Mater. 2017, 2, 224–247. [Google Scholar] [CrossRef] [PubMed]
  4. Habibovic, P.; Barralet, J.E. Bioinorganics and biomaterials: Bone repair. Acta Biomater. 2011, 7, 3013–3026. [Google Scholar] [CrossRef] [PubMed]
  5. van Rijt, S.H.; Sadler, P.J. Current applications and future potential for bioinorganic chemistry in the development of anticancer drugs. Drug Discov. Today 2009, 14, 1089–1097. [Google Scholar] [CrossRef] [Green Version]
  6. Ahmad, S.; Isab, A.A.; Ali, S.; Al-Arfaj, A.R. Perspectives in bioinorganic chemistry of some metal based therapeutic agents. Polyhedron 2006, 25, 1633–1645. [Google Scholar] [CrossRef]
  7. Cohen, S.M. New approaches for medicinal applications of bioinorganic chemistry. Curr. Opin. Chem. Biol. 2007, 11, 115–120. [Google Scholar] [CrossRef]
  8. Bouler, J.M.; Pilet, P.; Gauthier, O.; Verron, E. Biphasic calcium phosphate ceramics for bone reconstruction: A review of biological response. Acta Biomater 2017, 53, 1–12. [Google Scholar] [CrossRef]
  9. Barrere, F.; van Blitterswijk, C.A.; de Groot, K. Bone regeneration: Molecular and cellular interactions with calcium phosphate ceramics. Int. J. Nanomed. 2006, 1, 317–332. [Google Scholar]
  10. Bose, S.; Tarafder, S. Calcium phosphate ceramic systems in growth factor and drug delivery for bone tissue engineering: A review. Acta Biomater. 2012, 8, 1401–1421. [Google Scholar] [CrossRef] [Green Version]
  11. Wahl, D.A.; Czernuszka, J.T. Collagen-hydroxyapatite composites for hard tissue repair. Eur. Cells Mater. 2006, 11, 43–56. [Google Scholar] [CrossRef]
  12. Xie, J.; Baumann, M.J.; McCabe, L.R. Osteoblasts respond to hydroxyapatite surfaces with immediate changes in gene expression. J. Biomed. Mater. Res. Part A 2004, 71, 108–117. [Google Scholar] [CrossRef]
  13. Hu, C.; Zilm, M.; Wei, M. Fabrication of intrafibrillar and extrafibrillar mineralized collagen/apatite scaffolds with a hierarchical structure. J. Biomed. Mater. Res. Part A 2016, 104, 1153–1161. [Google Scholar] [CrossRef]
  14. Bhatt, R.A.; Rozental, T.D. Bone Graft Substitutes. Hand Clin. 2012, 28, 457–468. [Google Scholar] [CrossRef]
  15. Ripamonti, U. Soluble osteogenic molecular signals and the induction of bone formation. Biomaterials 2006, 27, 807–822. [Google Scholar] [CrossRef]
  16. Legeros, R.Z. Calcium phosphate-based osteoinductive materials. Chem. Rev. 2008, 108, 4742. [Google Scholar] [CrossRef]
  17. Bose, S.; Fielding, G.; Tarafder, S.; Bandyopadhyay, A. Understanding of dopant-induced osteogenesis and angiogenesis in calcium phosphate ceramics. Trends Biotechnol. 2013, 31, 594–605. [Google Scholar] [CrossRef] [Green Version]
  18. Kular, J.; Tickner, J.; Chim, S.M.; Xu, J. An overview of the regulation of bone remodelling at the cellular level. Clin. Biochem. 2012, 45, 863–873. [Google Scholar] [CrossRef]
  19. Silver, I.A.; Murrills, R.J.; Etherington, D.J. Microelectrode studies on the acid microenvironment beneath adherent macrophages and osteoclasts. Exp. Cell Res. 1988, 175, 266–276. [Google Scholar] [CrossRef]
  20. Solheim, E. Growth factors in bone. Int. Orthop. 1998, 22, 410–416. [Google Scholar] [CrossRef] [Green Version]
  21. Ash, C.; Stone, R. A question of dose-Introduction. Science 2003, 300, 925. [Google Scholar] [CrossRef] [Green Version]
  22. Sansone, V.; Pagani, D.; Melato, M. The effects on bone cells of metal ions released from orthopaedic implants: A review. Clin. Cases Miner. Bone Metab. 2013, 10, 34. [Google Scholar] [CrossRef] [PubMed]
  23. Thompson, K.; Orvig, C. Boon and bane of metal ions in medicine. Science 2003, 300, 936–939. [Google Scholar] [CrossRef] [PubMed]
  24. Olszta, M.J.; Cheng, X.; Jee, S.S.; Kumar, R.; Kim, Y.-Y.; Kaufman, M.J.; Douglas, E.P.; Gower, L.B. Bone structure and formation: A new perspective. Mater. Sci. Eng. R 2007, 58, 77–116. [Google Scholar] [CrossRef]
  25. Ma, G.; Liu, X.Y. Hydroxyapatite: Hexagonal or monoclinic? Cryst. Growth Des. 2009, 9, 2991–2994. [Google Scholar] [CrossRef]
  26. Morgan, H.; Wilson, R.M.; Elliott, J.C.; Dowker, S.E.P.; Anderson, P. Preparation and characterisation of monoclinic hydroxyapatite and its precipitated carbonate apatite intermediate. Biomaterials 2000, 21, 617–627. [Google Scholar] [CrossRef]
  27. Buddhachat, K.; Klinhom, S.; Siengdee, P.; Brown, J.L.; Nomsiri, R.; Kaewmong, P.; Thitaram, C.; Mahakkanukrauh, P.; Nganvongpanit, K. Elemental Analysis of Bone, Teeth, Horn and Antler in Different Animal Species Using Non-Invasive Handheld X-Ray Fluorescence. PLoS ONE 2016, 11, e0155458. [Google Scholar] [CrossRef]
  28. Castro, W.; Hoogewerff, J.; Latkoczy, C.; Almirall, J.R. Application of laser ablation (LA-ICP-SF-MS) for the elemental analysis of bone and teeth samples for discrimination purposes. Forensic Sci. Int. 2010, 195, 17–27. [Google Scholar] [CrossRef]
  29. Medvecký, Ľ.; Štulajterová, R.; Parilák, Ľ.; Trpčevská, J.; Ďurišin, J.; Barinov, S.M. Influence of manganese on stability and particle growth of hydroxyapatite in simulated body fluid. Colloids Surf. A Physicochem. Eng. Asp. 2006, 281, 221–229. [Google Scholar] [CrossRef]
  30. Zhang, J.; Dai, C.; Wei, J.; Wen, Z.; Zhang, S.; Lin, L. Calcium phosphate/chitosan composite coating: Effect of different concentrations of Mg2+ in the m-SBF on its bioactivity. Appl. Surf. Sci. 2013, 280, 256–262. [Google Scholar] [CrossRef]
  31. Rey, C.; Combes, C.; Drouet, C.; Glimcher, M. Bone mineral: Update on chemical composition and structure. Osteoporos. Int. 2009, 20, 1013–1021. [Google Scholar] [CrossRef] [Green Version]
  32. Abou Neel, E.A.; Aljabo, A.; Strange, A.; Ibrahim, S.; Coathup, M.; Young, A.M.; Bozec, L.; Mudera, V. Demineralization-remineralization dynamics in teeth and bone. Int. J. Nanomed. 2016, 11, 4743–4763. [Google Scholar] [CrossRef]
  33. Omelon, S.J.; Grynpas, M.D. Relationships between Polyphosphate Chemistry, Biochemistry and Apatite Biomineralization. Chem. Rev. 2008, 108, 4694–4715. [Google Scholar] [CrossRef]
  34. Wang, L.; Nancollas, G.H. Calcium Orthophosphates: Crystallization and Dissolution. Chem. Rev. 2008, 108, 4628–4669. [Google Scholar] [CrossRef] [Green Version]
  35. Best, S.M.; Porter, A.E.; Thian, E.S.; Huang, J. Bioceramics: Past, present and for the future. J. Eur. Ceram. Soc. 2008, 28, 1319–1327. [Google Scholar] [CrossRef]
  36. Dorozhkin, S.V.; Epple, M. Biological and Medical Significance of Calcium Phosphates. Angew. Chem. Int. Ed. Engl. 2002, 41, 3130–3146. [Google Scholar] [CrossRef]
  37. Albee, F.H. Studies in bone growth–triple calcium phosphate as stimulus to osteogenesis. Ann. Surg. 1920, 71, 32. [Google Scholar] [CrossRef]
  38. Roberts, S.C.; Brilliant, J.D. Tricalcium phosphate as an adjunct to apical closure in pulpless permanent teeth. J. Endod. 1975, 1, 263–269. [Google Scholar] [CrossRef]
  39. Nery, E.; Lynch, K.; Hirthe, W.; Mueller, K. Bioceramic implants in surgically produced infrabony defects. J. Periodontol. 1975, 46, 328–347. [Google Scholar] [CrossRef]
  40. Köster, K.; Karbe, E.; Kramer, H.; Heide, H.; König, R. Experimental bone replacement with resorbable calcium phosphate ceramic (author’s transl). Langenbecks Arch. Fur. Chir. 1976, 341, 77–86. [Google Scholar] [CrossRef] [PubMed]
  41. Denissen, H.W.; Groot, K.d. Immediate dental root implants from synthetic dense calcium hydroxylapatite. J. Prosthet. Dent. 1979, 42, 551–556. [Google Scholar] [CrossRef] [PubMed]
  42. Hofmann, G.; Kirschner, M.; Wangemann, T.; Falk, C.; Mempel, W.; Hammer, C. Infections and immunological hazards of allogeneic bone transplantation. Arch. Orthop. Trauma Surg. 1995, 114, 159–166. [Google Scholar] [CrossRef] [PubMed]
  43. Wenz, B.; Oesch, B.; Horst, M. Analysis of the risk of transmitting bovine spongiform encephalopathy through bone grafts derived from bovine bone. Biomaterials 2001, 22, 1599. [Google Scholar] [CrossRef]
  44. Hench, L.L.; Splinter, R.J.; Allen, W.C.; Greenlee, T.K. Bonding mechanisms at the interface of ceramic prosthetic materials. J. Biomed. Mater. Res. 1971, 5, 117–141. [Google Scholar] [CrossRef]
  45. Elsinger, E.C.; Leal, L. Coralline hydroxyapatite bone graft substitutes. J. Foot Ankle Surg. 1996, 35, 396–399. [Google Scholar] [CrossRef]
  46. Damien, E.; Revell, P.A. Coralline hydroxyapatite bone graft substitute: A review of experimental studies and biomedical applications. J. Appl. Biomater. Biomech. JABB 2004, 2, 65. [Google Scholar]
  47. Chou, J.; Austin, C.; Doble, P.; Ben-Nissan, B.; Milthorpe, B. Trace elemental imaging of coralline hydroxyapatite by laser-ablation inductively coupled plasma–mass spectroscopy. J. Tissue Eng. Regen. Med. 2014, 8, 515–520. [Google Scholar] [CrossRef]
  48. Yang, F.; Yang, D.; Tu, J.; Zheng, Q.; Cai, L.; Wang, L. Strontium Enhances Osteogenic Differentiation of Mesenchymal Stem Cells and In vivo Bone Formation by Activating Wnt/Catenin Signaling. Stem Cells 2011, 29, 981–991. [Google Scholar] [CrossRef]
  49. Ying, X.; Cheng, S.; Wang, W.; Lin, Z.; Chen, Q.; Zhang, W.; Kou, D.; Shen, Y.; Cheng, X.; Rompis, F.A.; et al. Effect of Boron on Osteogenic Differentiation of Human Bone Marrow Stromal Cells. Biol. Trace Elem. Res. 2011, 144, 306–315. [Google Scholar] [CrossRef]
  50. Meleti, Z.; Shapiro, I.M.; Adams, C.S. Inorganic phosphate induces apoptosis of osteoblast-like cells in culture. Bone 2000, 27, 359–366. [Google Scholar] [CrossRef]
  51. Mladenović, Ž.; Johansson, A.; Willman, B.; Shahabi, K.; Björn, E.; Ransjö, M. Soluble silica inhibits osteoclast formation and bone resorption in vitro. Acta Biomater. 2014, 10, 406–418. [Google Scholar] [CrossRef]
  52. Yamaguchi, M. Nutritional factors and bone homeostasis: Synergistic effect with zinc and genistein in osteogenesis. Mol. Cell. Biochem. 2012, 366, 201–221. [Google Scholar] [CrossRef]
  53. Verron, E.; Loubat, A.; Carle, G.F.; Vignes-Colombeix, C.; Strazic, I.; Guicheux, J.; Rochet, N.; Bouler, J.M.; Scimeca, J.-C. Molecular effects of gallium on osteoclastic differentiation of mouse and human monocytes. Biochem. Pharmacol. 2012, 83, 671–679. [Google Scholar] [CrossRef]
  54. Samavedi, S.; Whittington, A.R.; Goldstein, A.S. Calcium phosphate ceramics in bone tissue engineering: A review of properties and their influence on cell behavior. Acta Biomater. 2013, 9, 8037–8045. [Google Scholar] [CrossRef]
  55. Kasuya, S.; Kato-Kogoe, N.; Omori, M.; Yamamoto, K.; Taguchi, S.; Fujita, H.; Imagawa, N.; Sunano, A.; Inoue, K.; Ito, Y.; et al. New Bone Formation Process Using Bio-Oss and Collagen Membrane for Rat Calvarial Bone Defect: Histological Observation. Implant Dent. 2018, 27, 158–164. [Google Scholar] [CrossRef]
  56. Duda, M.; Pajak, J. The issue of bioresorption of the Bio-Oss xenogeneic bone substitute in bone defects. Ann. Univ. Mariae Curie Sklodowska Med. 2004, 59, 269–277. [Google Scholar]
  57. Kokubo, T.; Takadama, H. How useful is SBF in predicting in vivo bone bioactivity? Biomaterials 2006, 27, 2907–2915. [Google Scholar] [CrossRef]
  58. Sadat-Shojai, M.; Khorasani, M.-T.; Dinpanah-Khoshdargi, E.; Jamshidi, A. Synthesis methods for nanosized hydroxyapatite with diverse structures. Acta Biomater. 2013, 9, 7591–7621. [Google Scholar] [CrossRef]
  59. Zhou, Y.; Wu, C.; Chang, J. Bioceramics to regulate stem cells and their microenvironment for tissue regeneration. Mater. Today 2019, 24, 41–56. [Google Scholar] [CrossRef]
  60. Urist, M.R. Bone: Formation by Autoinduction. Science 1965, 150, 893. [Google Scholar] [CrossRef] [PubMed]
  61. Urist, M.R.; Strates, B.S. Bone Morphogenetic Protein. J. Dent. Res. 1971, 50, 1392–1406. [Google Scholar] [CrossRef] [PubMed]
  62. Hettiaratchi, M.H.; Krishnan, L.; Rouse, T.; Chou, C.; McDevitt, T.C.; Guldberg, R.E. Heparin-mediated delivery of bone morphogenetic protein-2 improves spatial localization of bone regeneration. Sci. Adv. 2020, 6, eaay1240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Urist, M.R.; Lietze, A.; Dawson, E. Beta-tricalcium phosphate delivery system for bone morphogenetic protein. Clin. Orthop. Relat. Res. 1984, 187, 277–280. [Google Scholar] [CrossRef]
  64. Wang, E.A.; Rosen, V.; Alessandro, J.S.; Bauduy, M.; Cordes, P.; Harada, T.; Israel, D.I.; Hewick, R.M.; Kerns, K.M.; LaPan, P. Recombinant human bone morphogenetic protein induces bone formation. Proc. Natl. Acad. Sci. USA 1990, 87, 2220–2224. [Google Scholar] [CrossRef] [Green Version]
  65. Wozney, J.; Rosen, V. Bone morphogenetic protein and bone morphogenetic protein gene family in bone formation and repair. Clin. Orthop. Relat. Res. 1998, 346, 26–37. [Google Scholar] [CrossRef]
  66. Boix, T.; Gómez-Morales, J.; Torrent-Burgués, J.; Monfort, A.; Puigdomènech, P.; Rodríguez-Clemente, R. Adsorption of recombinant human bone morphogenetic protein rhBMP-2m onto hydroxyapatite. J. Inorg. Biochem. 2005, 99, 1043–1050. [Google Scholar] [CrossRef]
  67. Einhorn, T.A.; Gerstenfeld, L.C. Fracture healing: Mechanisms and interventions. Nat. Rev. Rheumatol. 2015, 11, 45–54. [Google Scholar] [CrossRef] [Green Version]
  68. Kofron, M.D.; Laurencin, C.T. Bone tissue engineering by gene delivery. Adv. Drug Deliv. Rev. 2006, 58, 555–576. [Google Scholar] [CrossRef]
  69. Chattopadhyay, N.; Yano, S.; Tfelt-Hansen, J.; Rooney, P.; Kanuparthi, D.; Bandyopadhyay, S.; Ren, X.; Terwilliger, E.; Brown, E.M. Mitogenic Action of Calcium-Sensing Receptor on Rat Calvarial Osteoblasts. Endocrinology 2004, 145, 3451–3462. [Google Scholar] [CrossRef] [Green Version]
  70. Malhotra, A.; Habibovic, P. Calcium Phosphates and Angiogenesis: Implications and Advances for Bone Regeneration. Trends Biotechnol. 2016, 34, 983–992. [Google Scholar] [CrossRef] [Green Version]
  71. Kokubo, T. Bioactive glass ceramics: Properties and applications. Biomaterials 1991, 12, 155–163. [Google Scholar] [CrossRef]
  72. Bohner, M.; Lemaitre, J. Can bioactivity be tested in vitro with SBF solution? Biomaterials 2009, 30, 2175–2179. [Google Scholar] [CrossRef] [Green Version]
  73. Pan, H.; Zhao, X.; Darvell, B.W.; Lu, W.W. Apatite-formation ability–predictor of “bioactivity”? Acta Biomater. 2010, 6, 4181–4188. [Google Scholar] [CrossRef]
  74. Dermience, M.; Lognay, G.; Mathieu, F.; Goyens, P. Effects of thirty elements on bone metabolism. J. Trace Elem. Med. Biol. 2015, 32, 86–106. [Google Scholar] [CrossRef]
  75. Khoshniat, S.; Bourgine, A.; Julien, M.; Weiss, P.; Guicheux, J.; Beck, L. The emergence of phosphate as a specific signaling molecule in bone and other cell types in mammals. Cell. Mol. Life Sci. 2011, 68, 205–218. [Google Scholar] [CrossRef]
  76. Liu, Y.K.; Lu, Q.Z.; Pei, R.; Ji, H.J.; Zhou, G.S.; Zhao, X.L.; Tang, R.K.; Zhang, M. The effect of extracellular calcium and inorganic phosphate on the growth and osteogenic differentiation of mesenchymal stem cells in vitro: Implication for bone tissue engineering. Biomed. Mater. 2009, 4, 025004. [Google Scholar] [CrossRef]
  77. Huang, Z.; Cheng, S.L.; Slatopolsky, E. Sustained activation of the extracellular signal-regulated kinase pathway is required for extracellular calcium stimulation of human osteoblast proliferation. J. Biol. Chem. 2001, 276, 21351. [Google Scholar] [CrossRef] [Green Version]
  78. Fromigué, O.; Haÿ, E.; Barbara, A.; Petrel, C.; Traiffort, E.; Ruat, M.; Marie, P.J. Calcium sensing receptor-dependent and receptor-independent activation of osteoblast replication and survival by strontium ranelate. J. Cell. Mol. Med. 2009, 13, 2189–2199. [Google Scholar] [CrossRef]
  79. Mentaverri, R.; Yano, S.; Chattopadhyay, N.; Petit, L.; Kifor, O.; Kamel, S.; Terwilliger, E.F.; Brazier, M.; Brown, E.M. The calcium sensing receptor is directly involved in both osteoclast differentiation and apoptosis. FASEB J. 2006, 20, 2562–2564. [Google Scholar] [CrossRef]
  80. Zaidi, M.; Adebanjo, O.A.; Moonga, B.S.; Sun, L.; Huang, C.L.H. Emerging Insights into the Role of Calcium Ions in Osteoclast Regulation. J. Bone Miner. Res. 1999, 14, 669–674. [Google Scholar] [CrossRef] [PubMed]
  81. Habibovic, P.; Bassett, D.C.; Doillon, C.J.; Gerard, C.; McKee, M.D.; Barralet, J.E. Collagen biomineralization in vivo by sustained release of inorganic phosphate ions. Adv. Mater. 2010, 22, 1858–1862. [Google Scholar] [CrossRef] [PubMed]
  82. Yoshiko, Y.; Candeliere, G.A.; Maeda, N.; Aubin, J.E. Osteoblast autonomous Pi regulation via Pit1 plays a role in bone mineralization. Mol. Cell. Biol. 2007, 27, 4465–4474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Adams, C.S.; Mansfield, K.; Perlot, R.L.; Shapiro, I.M. Matrix regulation of skeletal cell apoptosis. Role of calcium and phosphate ions. J. Biol. Chem. 2001, 276, 20316–20322. [Google Scholar] [CrossRef] [Green Version]
  84. Kanatani, M.; Sugimoto, T.; Kano, J.; Kanzawa, M.; Chihara, K. Effect of high phosphate concentration on osteoclast differentiation as well as bone-resorbing activity. J. Cell. Physiol. 2003, 196, 180–189. [Google Scholar] [CrossRef]
  85. Mozar, A.; Haren, N.; Chasseraud, M.; Louvet, L.; Mazière, C.; Wattel, A.; Mentaverri, R.; Morlière, P.; Kamel, S.; Brazier, M. High extracellular inorganic phosphate concentration inhibits RANK–RANKL signaling in osteoclast-like cells. J. Cell. Physiol. 2008, 215, 47–54. [Google Scholar] [CrossRef]
  86. Brown, R.B.; Razzaque, M.S. Dysregulation of phosphate metabolism and conditions associated with phosphate toxicity. Bonekey Rep. 2015, 4, 705. [Google Scholar] [CrossRef] [Green Version]
  87. Nielsen, F.H. Dietary fat composition modifies the effect of boron on bone characteristics and plasma lipids in rats. Biofactors 2004, 20, 161–171. [Google Scholar] [CrossRef]
  88. Gorustovich, A.A.; Steimetz, T.; Nielsen, F.H.; Guglielmotti, M.B. Histomorphometric study of alveolar bone healing in rats fed a boron-deficient diet. Anat. Rec. 2008, 291, 441–447. [Google Scholar] [CrossRef]
  89. Nielsen, F.H.; Stoecker, B.J. Boron and fish oil have different beneficial effects on strength and trabecular microarchitecture of bone. J. Trace Elem. Med. Biol. 2009, 23, 195–203. [Google Scholar] [CrossRef]
  90. Uysal, T.; Ustdal, A.; Sonmez, M.F.; Ozturk, F. Stimulation of Bone Formation by Dietary Boron in an Orthopedically Expanded Suture in Rabbits. Angle Orthod. 2009, 79, 984–990. [Google Scholar] [CrossRef] [Green Version]
  91. Movahedi Najafabadi, B.-A.-H.; Abnosi, M.H. Boron Induces Early Matrix Mineralization via Calcium Deposition and Elevation of Alkaline Phosphatase Activity in Differentiated Rat Bone Marrow Mesenchymal Stem Cells. Cell J. 2016, 18, 62–73. [Google Scholar] [CrossRef]
  92. Hakki, S.S.; Bozkurt, B.S.; Hakki, E.E. Boron regulates mineralized tissue-associated proteins in osteoblasts (MC3T3-E1). J. Trace Elem. Med. Biol. 2010, 24, 243–250. [Google Scholar] [CrossRef]
  93. Roughead, Z.K.; Lukaski, H.C. Inadequate Copper Intake Reduces Serum Insulin-Like Growth Factor-I and Bone Strength in Growing Rats Fed Graded Amounts of Copper and Zinc. J. Nutr. 2003, 133, 442–448. [Google Scholar] [CrossRef] [Green Version]
  94. Rucker, R.B.; Kosonen, T.; Clegg, M.S.; Mitchell, A.E.; Rucker, B.R.; Uriu-Hare, J.Y.; Keen, C.L. Copper, lysyl oxidase, and extracellular matrix protein cross-linking. Am. J. Clin. Nutr. 1998, 67, 996S–1002S. [Google Scholar] [CrossRef] [Green Version]
  95. Uauy, R.; Maass, A.; Araya, M. Estimating risk from copper excess in human populations. Am. J. Clin. Nutr. 2008, 88, 867S–871S. [Google Scholar] [CrossRef] [Green Version]
  96. Barralet, J.; Gbureck, U.; Habibovic, P.; Vorndran, E.; Gerard, C.; Doillon, C.J. Angiogenesis in calcium phosphate scaffolds by inorganic copper ion release. Tissue Eng. Part A 2009, 15, 1601–1609. [Google Scholar] [CrossRef]
  97. Wu, C.; Zhou, Y.; Xu, M.; Han, P.; Chen, L.; Chang, J.; Xiao, Y. Copper-containing mesoporous bioactive glass scaffolds with multifunctional properties of angiogenesis capacity, osteostimulation and antibacterial activity. Biomaterials 2013, 34, 422–433. [Google Scholar] [CrossRef]
  98. Neel, E.A.A.; Ahmed, I.; Pratten, J.; Nazhat, S.N.; Knowles, J.C. Characterisation of antibacterial copper releasing degradable phosphate glass fibres. Biomaterials 2005, 26, 2247–2254. [Google Scholar] [CrossRef]
  99. Finney, L.; Mandava, S.; Ursos, L.; Zhang, W.; Rodi, D.; Vogt, S.; Legnini, D.; Maser, J.; Ikpatt, F.; Olopade, O.I. X-ray fluorescence microscopy reveals large-scale relocalization and extracellular translocation of cellular copper during angiogenesis. Proc. Natl. Acad. Sci. USA 2007, 104, 2247–2252. [Google Scholar] [CrossRef] [Green Version]
  100. Verron, E.; Bouler, J.M.; Scimeca, J.C. Gallium as a potential candidate for treatment of osteoporosis. Drug Discov. Today 2012, 17, 1127–1132. [Google Scholar] [CrossRef] [PubMed]
  101. Bockman, R. The effects of gallium nitrate on bone resorption. Semin. Oncol. 2003, 2003, 5–12. [Google Scholar] [CrossRef] [PubMed]
  102. Hall, T.J.; Chambers, T.J. Gallium inhibits bone resorption by a direct effect on osteoclasts. Bone Miner. 1990, 8, 211–216. [Google Scholar] [CrossRef] [PubMed]
  103. Rude, R.K.; Gruber, H.E.; Norton, H.J.; Wei, L.Y.; Frausto, A.; Kilburn, J. Reduction of dietary magnesium by only 50% in the rat disrupts bone and mineral metabolism. Osteoporos. Int. 2006, 17, 1022–1032. [Google Scholar] [CrossRef] [PubMed]
  104. Rude, R.K.; Gruber, H.E.; Norton, H.J.; Wei, L.Y.; Frausto, A.; Kilburn, J. Dietary magnesium reduction to 25% of nutrient requirement disrupts bone and mineral metabolism in the rat. Bone 2005, 37, 211–219. [Google Scholar] [CrossRef]
  105. Rude, R.K.; Singer, F.R.; Gruber, H.E. Skeletal and hormonal effects of magnesium deficiency. J. Am. Coll. Nutr. 2009, 28, 131–141. [Google Scholar] [CrossRef]
  106. Castiglioni, S.; Cazzaniga, A.; Albisetti, W.; Maier, A.M.J. Magnesium and Osteoporosis: Current State of Knowledge and Future Research Directions. Nutrients 2013, 5, 3022. [Google Scholar] [CrossRef] [Green Version]
  107. Rude, R.K.; Gruber, H.E.; Wei, L.Y.; Frausto, A.; Mills, B.G. Magnesium Deficiency: Effect on Bone and Mineral Metabolism in the Mouse. Calcif. Tissue Int. 2003, 72, 32–41. [Google Scholar] [CrossRef]
  108. Yoshizawa, S.; Brown, A.; Barchowsky, A.; Sfeir, C. Magnesium ion stimulation of bone marrow stromal cells enhances osteogenic activity, simulating the effect of magnesium alloy degradation. Acta Biomater. 2014, 10, 2834–2842. [Google Scholar] [CrossRef]
  109. Witte, F.; Kaese, V.; Haferkamp, H.; Switzer, E.; Meyer-Lindenberg, A.; Wirth, C.J.; Windhagen, H. In vivo corrosion of four magnesium alloys and the associated bone response. Biomaterials 2005, 26, 3557–3563. [Google Scholar] [CrossRef]
  110. Galli, S.; Naito, Y.; Karlsson, J.; He, W.; Miyamoto, I.; Xue, Y.; Andersson, M.; Mustafa, K.; Wennerberg, A.; Jimbo, R. Local release of magnesium from mesoporous TiO2 coatings stimulates the peri-implant expression of osteogenic markers and improves osteoconductivity in vivo. Acta Biomater. 2014, 10, 5193–5201. [Google Scholar] [CrossRef]
  111. Leidi, M.; Dellera, F.; Mariotti, M.; Maier, J.A.M. High magnesium inhibits human osteoblast differentiation in vitro. Magnes. Res. 2011, 24, 1–6. [Google Scholar] [CrossRef]
  112. Navarro-González, J.F.; Mora-Fernández, C.; García-Pérez, J. Clinical implications of disordered magnesium homeostasis in chronic renal failure and dialysis. Semin. Dial. 2009, 22, 37–44. [Google Scholar] [CrossRef]
  113. Katsumata, S.-i.; Katsumata-Tsuboi, R.; Uehara, M.; Suzuki, K. Severe Iron Deficiency Decreases Both Bone Formation and Bone Resorption in Rats. J. Nutr. 2008, 139, 238–243. [Google Scholar] [CrossRef] [Green Version]
  114. Tsay, J.; Yang, Z.; Ross, F.P.; Cunningham-Rundles, S.; Lin, H.; Coleman, R.; Mayer-Kuckuk, P.; Doty, S.B.; Grady, R.W.; Giardina, P.J.; et al. Bone loss caused by iron overload in a murine model: Importance of oxidative stress. Blood 2010, 116, 2582–2589. [Google Scholar] [CrossRef] [Green Version]
  115. Isomura, H.; Fujie, K.; Shibata, K.; Inoue, N.; Iizuka, T.; Takebe, G.; Takahashi, K.; Nishihira, J.; Izumi, H.; Sakamoto, W. Bone metabolism and oxidative stress in postmenopausal rats with iron overload. Toxicology 2004, 197, 92–99. [Google Scholar] [CrossRef]
  116. He, Y.-F.; Ma, Y.; Gao, C.; Zhao, G.-y.; Zhang, L.-L.; Li, G.-F.; Pan, Y.-Z.; Li, K.; Xu, Y.-J. Iron Overload Inhibits Osteoblast Biological Activity Through Oxidative Stress. Biol. Trace Elem. Res. 2013, 152, 292–296. [Google Scholar] [CrossRef]
  117. Beattie, J.H.; Avenell, A. Trace element nutrition and bone metabolism. Nutr. Res. Rev. 1992, 5, 167–188. [Google Scholar] [CrossRef]
  118. O’Neal, S.L.; Zheng, W. Manganese toxicity upon overexposure: A decade in review. Curr. Environ. Health Rep. 2015, 2, 315–328. [Google Scholar] [CrossRef] [Green Version]
  119. Vieira, S.I.; Cerqueira, A.R.; Pina, S.; da Cruz Silva, O.A.B.; Abrantes, J.C.C.; Ferreira, J.M.F. Effects of Mn-doping on the structure and biological properties of β-tricalcium phosphate. J. Inorg. Biochem. 2014, 136, 57–66. [Google Scholar]
  120. Miola, M.; Brovarone, C.V.; Maina, G.; Rossi, F.; Bergandi, L.; Ghigo, D.; Saracino, S.; Maggiora, M.; Canuto, R.A.; Muzio, G. In vitro study of manganese-doped bioactive glasses for bone regeneration. Mater. Sci. Eng. C 2014, 38, 107–118. [Google Scholar] [CrossRef] [PubMed]
  121. Barrioni, B.R.; Norris, E.; Li, S.; Naruphontjirakul, P.; Jones, J.R.; de Magalhães Pereira, M. Osteogenic potential of sol–gel bioactive glasses containing manganese. J. Mater. Sci. Mater. Med. 2019, 30, 86. [Google Scholar] [CrossRef]
  122. Yu, L.; Tian, Y.; Qiao, Y.; Liu, X. Mn-containing titanium surface with favorable osteogenic and antimicrobial functions synthesized by PIII&D. Colloids Surf. B Biointerfaces 2017, 152, 376–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Hreha, J.; Wey, A.; Cunningham, C.; Krell, E.S.; Brietbart, E.A.; Paglia, D.N.; Montemurro, N.J.; Nguyen, D.A.; Lee, Y.-J.; Komlos, D.; et al. Local manganese chloride treatment accelerates fracture healing in a rat model. J. Orthop. Res. 2015, 33, 122–130. [Google Scholar] [CrossRef] [PubMed]
  124. Downey, C.M.; Horton, C.R.; Carlson, B.A.; Parsons, T.E.; Hatfield, D.L.; Hallgrímsson, B.; Jirik, F.R. Osteo-chondroprogenitor–specific deletion of the selenocysteine trna gene, Trsp, leads to chondronecrosis and abnormal skeletal development: A putative model for kashin-beck disease. PLoS Genet. 2009, 5, e1000616. [Google Scholar] [CrossRef] [Green Version]
  125. Moreno-Reyes, R.; Egrise, D.; Nève, J.; Pasteels, J.L.; Schoutens, A. Selenium deficiency-induced growth retardation is associated with an impaired bone metabolism and osteopenia. J. Bone Miner. Res. 2001, 16, 1556–1563. [Google Scholar] [CrossRef]
  126. Ren, F.L.; Guo, X.; Zhang, R.J.; Wang, S.J.; Zuo, H.; Zhang, Z.T.; Geng, D.; Yu, Y.; Su, M. Effects of selenium and iodine deficiency on bone, cartilage growth plate and chondrocyte differentiation in two generations of rats. Osteoarthr. Cartil. 2007, 15, 1171–1177. [Google Scholar] [CrossRef] [Green Version]
  127. Yang, C.; Wolf, E.; Röser, K.; Delling, G.; Müller, P.K. Selenium deficiency and fulvic acid supplementation induces fibrosis of cartilage and disturbs subchondral ossification in knee joints of mice: An animal model study of Kashin-Beck disease. Virchows Arch. A 1993, 423, 483–491. [Google Scholar] [CrossRef]
  128. Martiniaková, M.; Boboňová, I.; Omelka, R.; Grosskopf, B.; Stawarz, R.; Toman, R. Structural changes in femoral bone tissue of rats after subchronic peroral exposure to selenium. Acta Vet. Scand. 2013, 55, 8. [Google Scholar] [CrossRef] [Green Version]
  129. Turan, B.; Bayari, S.; Balcik, C.; Severcan, F.; Akkas, N. A biomechanical and spectroscopic study of bone from rats with selenium deficiency and toxicity. Biometals 2000, 13, 113–121. [Google Scholar] [CrossRef]
  130. Hoeg, A.; Gogakos, A.; Murphy, E.; Mueller, S.; Köhrle, J.; Reid, D.M.; Glüer, C.C.; Felsenberg, D.; Roux, C.; Eastell, R.; et al. Bone Turnover and Bone Mineral Density Are Independently Related to Selenium Status in Healthy Euthyroid Postmenopausal Women. J. Clin. Endocrinol. Metab. 2012, 97, 4061–4070. [Google Scholar] [CrossRef] [Green Version]
  131. McCann, J.C.; Ames, B.N. Adaptive dysfunction of selenoproteins from the perspective of the triage theory: Why modest selenium deficiency may increase risk of diseases of aging. FASEB J. 2011, 25, 1793–1814. [Google Scholar] [CrossRef] [Green Version]
  132. Xynos, I.D.; Edgar, A.J.; Buttery, L.D.K.; Hench, L.L.; Polak, J.M. Ionic Products of Bioactive Glass Dissolution Increase Proliferation of Human Osteoblasts and Induce Insulin-like Growth Factor II mRNA Expression and Protein Synthesis. Biochem. Biophys. Res. Commun. 2000, 276, 461–465. [Google Scholar] [CrossRef]
  133. Xynos, I.D.; Hukkanen, M.V.J.; Batten, J.J.; Buttery, L.D.; Hench, L.L.; Polak, J.M. Bioglass ®45S5 Stimulates Osteoblast Turnover and Enhances Bone Formation In vitro: Implications and Applications for Bone Tissue Engineering. Calcif. Tissue Int. 2000, 67, 321–329. [Google Scholar] [CrossRef]
  134. Hench, L.L.; Jones, J.R. Bioactive Glasses: Frontiers and Challenges. Front. Bioeng. Biotechnol. 2015, 3, 194. [Google Scholar] [CrossRef] [Green Version]
  135. Reffitt, D.M.; Ogston, N.; Jugdaohsingh, R.; Cheung, H.F.J.; Evans, B.A.J.; Thompson, R.P.H.; Powell, J.J.; Hampson, G.N. Orthosilicic acid stimulates collagen type 1 synthesis and osteoblastic differentiation in human osteoblast-like cells in vitro. Bone 2003, 32, 127–135. [Google Scholar] [CrossRef]
  136. Han, P.; Wu, C.; Xiao, Y. The effect of silicate ions on proliferation, osteogenic differentiation and cell signalling pathways (WNT and SHH) of bone marrow stromal cells. Biomater. Sci. 2013, 1, 379–392. [Google Scholar] [CrossRef]
  137. Zhou, X.; Moussa, F.M.; Mankoci, S.; Ustriyana, P.; Zhang, N.; Abdelmagid, S.; Molenda, J.; Murphy, W.L.; Safadi, F.F.; Sahai, N. Orthosilicic acid, Si(OH)4, stimulates osteoblast differentiation in vitro by upregulating miR-146a to antagonize NF-κB activation. Acta Biomater. 2016, 39, 192–202. [Google Scholar] [CrossRef]
  138. Schröder, H.C.; Wang, X.H.; Wiens, M.; Diehl-Seifert, B.; Kropf, K.; Schloßmacher, U.; Müller, W.E.G. Silicate modulates the cross-talk between osteoblasts (SaOS-2) and osteoclasts (RAW 264.7 cells): Inhibition of osteoclast growth and differentiation. J. Cell. Biochem. 2012, 113, 3197–3206. [Google Scholar] [CrossRef]
  139. Henstock, J.R.; Ruktanonchai, U.R.; Canham, L.T.; Anderson, S.I. Porous silicon confers bioactivity to polycaprolactone composites in vitro. J. Mater. Sci. Mater. Med. 2014, 25, 1087–1097. [Google Scholar] [CrossRef]
  140. Shi, M.; Zhou, Y.; Shao, J.; Chen, Z.; Song, B.; Chang, J.; Wu, C.; Xiao, Y. Stimulation of osteogenesis and angiogenesis of hBMSCs by delivering Si ions and functional drug from mesoporous silica nanospheres. Acta Biomater. 2015, 21, 178–189. [Google Scholar] [CrossRef] [PubMed]
  141. Chattopadhyay, N.; Quinn, S.J.; Kifor, O.; Ye, C.; Brown, E.M. The calcium-sensing receptor (CaR) is involved in strontium ranelate-induced osteoblast proliferation. Biochem. Pharmacol. 2007, 74, 438–447. [Google Scholar] [CrossRef] [PubMed]
  142. Bonnelye, E.; Chabadel, A.; Saltel, F.; Jurdic, P. Dual effect of strontium ranelate: Stimulation of osteoblast differentiation and inhibition of osteoclast formation and resorption in vitro. Bone 2008, 42, 129–138. [Google Scholar] [CrossRef] [PubMed]
  143. Peng, S.; Zhou, G.; Luk, K.D.K.; Cheung, K.M.C.; Li, Z.; Lam, W.M.; Zhou, Z.; Lu, W.W. Strontium Promotes Osteogenic Differentiation of Mesenchymal Stem Cells Through the Ras/MAPK Signaling Pathway. Cell. Physiol. Biochem. 2009, 23, 165–174. [Google Scholar] [CrossRef]
  144. Choudhary, S.; Halbout, P.; Alander, C.; Raisz, L.; Pilbeam, C. Strontium Ranelate Promotes Osteoblastic Differentiation and Mineralization of Murine Bone Marrow Stromal Cells: Involvement of Prostaglandins. J. Bone Miner. Res. 2007, 22, 1002–1010. [Google Scholar] [CrossRef]
  145. Marie, P.J. Strontium ranelate: A physiological approach for optimizing bone formation and resorption. Bone 2006, 38, 10–14. [Google Scholar] [CrossRef]
  146. Brennan, T.C.; Rybchyn, M.S.; Green, W.; Atwa, S.; Conigrave, A.D.; Mason, R.S. Osteoblasts play key roles in the mechanisms of action of strontium ranelate. Br. J. Pharmacol. 2009, 157, 1291–1300. [Google Scholar] [CrossRef] [Green Version]
  147. Wornham, D.P.; Hajjawi, M.O.; Orriss, I.R.; Arnett, T.R. Strontium potently inhibits mineralisation in bone-forming primary rat osteoblast cultures and reduces numbers of osteoclasts in mouse marrow cultures. Osteoporos. Int. 2014, 25, 2477–2484. [Google Scholar] [CrossRef]
  148. Yamaguchi, M.; Neale Weitzmann, M. The intact strontium ranelate complex stimulates osteoblastogenesis and suppresses osteoclastogenesis by antagonizing NF-κB activation. Mol. Cell. Biochem. 2012, 359, 399–407. [Google Scholar] [CrossRef]
  149. Peng, S.; Liu, X.S.; Huang, S.; Li, Z.; Pan, H.; Zhen, W.; Luk, K.D.K.; Guo, X.E.; Lu, W.W. The cross-talk between osteoclasts and osteoblasts in response to strontium treatment: Involvement of osteoprotegerin. Bone 2011, 49, 1290–1298. [Google Scholar] [CrossRef]
  150. Baron, R.; Tsouderos, Y. In vitro effects of S12911-2 on osteoclast function and bone marrow macrophage differentiation. Eur. J. Pharmacol. 2002, 450, 11–17. [Google Scholar] [CrossRef]
  151. Hurtel-Lemaire, A.S.; Mentaverri, R.; Caudrillier, A.; Cournarie, F.; Wattel, A.; Kamel, S.; Terwilliger, E.F.; Brown, E.M.; Brazier, M. The calcium-sensing receptor is involved in strontium ranelate-induced osteoclast apoptosis. New insights into the associated signaling pathways. J. Biol. Chem. 2009, 284, 575. [Google Scholar] [CrossRef] [Green Version]
  152. Zhang, W.; Wang, G.; Liu, Y.; Zhao, X.; Zou, D.; Zhu, C.; Jin, Y.; Huang, Q.; Sun, J.; Liu, X.; et al. The synergistic effect of hierarchical micro/nano-topography and bioactive ions for enhanced osseointegration. Biomaterials 2013, 34, 3184–3195. [Google Scholar] [CrossRef]
  153. Luo, X.; Barbieri, D.; Zhang, Y.; Yan, Y.; Bruijn, J.D.; Yuan, H. Strontium-Containing Apatite/Poly Lactide Composites Favoring Osteogenic Differentiation and in vivo Bone Formation. ACS Biomater. Sci. Eng. 2015, 1, 85–93. [Google Scholar] [CrossRef]
  154. Hill, T.; Meunier, N.; Andriollo-Sanchez, M.; Ciarapica, D.; Hininger-Favier, I.; Polito, A.; O’Connor, J.M.; Coudray, C.; Cashman, K.D. The relationship between the zinc nutritive status and biochemical markers of bone turnover in older European adults: The ZENITH study. Eur. J. Clin. Nutr. 2005, 59, S73–S78. [Google Scholar] [CrossRef] [Green Version]
  155. Rossi, L.; Migliaccio, S.; Corsi, A.; Marzia, M.; Bianco, P.; Teti, A.; Gambelli, L.; Cianfarani, S.; Paoletti, F.; Branca, F. Reduced growth and skeletal changes in zinc-deficient growing rats are due to impaired growth plate activity and inanition. J. Nutr. 2001, 131, 1142–1146. [Google Scholar] [CrossRef] [Green Version]
  156. Yusa, K.; Yamamoto, O.; Fukuda, M.; Koyota, S.; Koizumi, Y.; Sugiyama, T. In vitro prominent bone regeneration by release zinc ion from Zn-modified implant. Biochem. Biophys. Res. Commun. 2011, 412, 273–278. [Google Scholar] [CrossRef]
  157. Hoppe, A.; Güldal, N.S.; Boccaccini, A.R. A review of the biological response to ionic dissolution products from bioactive glasses and glass-ceramics. Biomaterials 2011, 32, 2757–2774. [Google Scholar] [CrossRef]
  158. Hadley, K.B.; Newman, S.M.; Hunt, J.R. Dietary zinc reduces osteoclast resorption activities and increases markers of osteoblast differentiation, matrix maturation, and mineralization in the long bones of growing rats. J. Nutr. Biochem. 2010, 21, 297–303. [Google Scholar] [CrossRef]
  159. Yamaguchi, M. Role of zinc in bone formation and bone resorption. J. Trace Elem. Exp. Med. Off. Publ. Int. Soc. Trace Elem. Res. Hum. 1998, 11, 119–135. [Google Scholar] [CrossRef]
  160. Kwun, I.-S.; Cho, Y.-E.; Lomeda, R.-A.R.; Shin, H.-I.; Choi, J.-Y.; Kang, Y.-H.; Beattie, J.H. Zinc deficiency suppresses matrix mineralization and retards osteogenesis transiently with catch-up possibly through Runx 2 modulation. Bone 2010, 46, 732–741. [Google Scholar] [CrossRef] [PubMed]
  161. Seo, H.-J.; Cho, Y.-E.; Kim, T.; Shin, H.-I.; Kwun, I.-S. Zinc may increase bone formation through stimulating cell proliferation, alkaline phosphatase activity and collagen synthesis in osteoblastic MC3T3-E1 cells. Nutr. Res. Pract. 2010, 4, 356–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Moonga, B.S.; Dempster, D.W. Zinc is a potent inhibitor of osteoclastic bone resorption in vitro. J. Bone Miner. Res. 1995, 10, 453–457. [Google Scholar] [CrossRef] [PubMed]
  163. Yamaguchi, M.; Weitzmann, M.N. Zinc stimulates osteoblastogenesis and suppresses osteoclastogenesis by antagonizing NF-κB activation. Mol. Cell. Biochem. 2011, 355, 179. [Google Scholar] [CrossRef] [PubMed]
  164. Marie, P.J. The calcium-sensing receptor in bone cells: A potential therapeutic target in osteoporosis. Bone 2010, 46, 571–576. [Google Scholar] [CrossRef]
  165. Melita, M.D.; Ashia, S.; Donald, T.W.; Carter, D.H.; Sarah, L.D.; Edward, F.N.; Daniela, R. Physiological changes in extracellular calcium concentration directly control osteoblast function in the absence of calciotropic hormones. Proc. Natl. Acad. Sci. USA 2004, 101, 5140. [Google Scholar] [CrossRef] [Green Version]
  166. Aguirre, A.; González, A.; Planell, J.A.; Engel, E. Extracellular calcium modulates in vitro bone marrow-derived Flk-1+ CD34+ progenitor cell chemotaxis and differentiation through a calcium-sensing receptor. Biochem. Biophys. Res. Commun. 2010, 393, 156–161. [Google Scholar] [CrossRef]
  167. Gustavsson, J.; Ginebra, M.; Planell, J.; Engel, E. Osteoblast-like cellular response to dynamic changes in the ionic extracellular environment produced by calcium-deficient hydroxyapatite. J. Mater. Sci. Mater. Med. 2012, 23, 2509–2520. [Google Scholar] [CrossRef]
  168. Yamauchi, M.; Yamaguchi, T.; Kaji, H.; Sugimoto, T.; Chihara, K. Involvement of calcium-sensing receptor in osteoblastic differentiation of mouse MC3T3-E1 cells. Am. J. Physiol.-Endocrinol. Metab. 2005, 288, E608–E616. [Google Scholar] [CrossRef] [Green Version]
  169. Chai, Y.C.; Roberts, S.J.; Schrooten, J.; Luyten, F.P. Probing the Osteoinductive Effect of Calcium Phosphate by Using an In vitro Biomimetic Model. Tissue Eng. Part A 2010, 17, 1083–1097. [Google Scholar] [CrossRef]
  170. Nakade, O.; Takahashi, K.; Takuma, T.; Aoki, T.; Kaku, T. Effect of extracellular calcium on the gene expression of bone morphogenetic protein-2 and -4 of normal human bone cells. J. Bone Miner. Metab. 2001, 19, 13–19. [Google Scholar] [CrossRef]
  171. Maeno, S.; Niki, Y.; Matsumoto, H.; Morioka, H.; Yatabe, T.; Funayama, A.; Toyama, Y.; Taguchi, T.; Tanaka, J. The effect of calcium ion concentration on osteoblast viability, proliferation and differentiation in monolayer and 3D culture. Biomaterials 2005, 26, 4847–4855. [Google Scholar] [CrossRef]
  172. Godwin, S.L.; Soltoff, S.P. Calcium-sensing receptor-mediated activation of phospholipase C-γ1 is downstream of phospholipase C-β and protein kinase C in MC3T3-E1 osteoblasts. Bone 2002, 30, 559–566. [Google Scholar] [CrossRef]
  173. Choudhary, S.; Kumar, A.; Kale, R.K.; Raisz, L.G.; Pilbeam, C.C. Extracellular calcium induces COX-2 in osteoblasts via a PKA pathway. Biochem. Biophys. Res. Commun. 2004, 322, 395–402. [Google Scholar] [CrossRef]
  174. Honda, Y.; Fitzsimmons, R.J.; Baylink, D.J.; Mohan, S. Effects of extracellular calcium on insulin-like growth factor II in human bone cells. J. Bone Miner. Res. 1995, 10, 1660–1665. [Google Scholar] [CrossRef]
  175. Choudhary, S.; Wadhwa, S.; Raisz, L.G.; Alander, C.; Pilbeam, C.C. Extracellular Calcium Is a Potent Inducer of Cyclo-oxygenase-2 in Murine Osteoblasts Through an ERK Signaling Pathway. J. Bone Miner. Res. 2003, 18, 1813–1824. [Google Scholar] [CrossRef]
  176. Nakamura, S.; Matsumoto, T.; Sasaki, J.-I.; Egusa, H.; Lee, K.Y.; Nakano, T.; Sohmura, T.; Nakahira, A. Effect of Calcium Ion Concentrations on Osteogenic Differentiation and Hematopoietic Stem Cell Niche-Related Protein Expression in Osteoblasts. Tissue Eng. Part A 2010, 16, 2467–2473. [Google Scholar] [CrossRef]
  177. Bergwitz, C.; Jüppner, H. Regulation of Phosphate Homeostasis by PTH, Vitamin D, and FGF23. Annu. Rev. Med. 2010, 61, 91–104. [Google Scholar] [CrossRef] [Green Version]
  178. Kanatani, M.; Sugimoto, T.; Kano, J.; Chihara, K. IGF-I mediates the stimulatory effect of high phosphate concentration on osteoblastic cell proliferation. J. Cell. Physiol. 2002, 190, 306–312. [Google Scholar] [CrossRef]
  179. Conrads, K.A.; Yi, M.; Simpson, K.A.; Lucas, D.A.; Camalier, C.E.; Yu, L.-R.; Veenstra, T.D.; Stephens, R.M.; Conrads, T.P.; Beck, G.R. A Combined Proteome and Microarray Investigation of Inorganic Phosphate-induced Pre-osteoblast Cells. Mol. Cell. Proteom. 2005, 4, 1284. [Google Scholar] [CrossRef] [Green Version]
  180. Beck, G.R.; Knecht, N. Osteopontin regulation by inorganic phosphate is ERK1/2-, protein kinase C-, and proteasome-dependent. J. Biol. Chem. 2003, 278, 41921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Julien, M.; Khoshniat, S.; Lacreusette, A.; Gatius, M.; Bozec, A.; Wagner, E.F.; Wittrant, Y.; Masson, M.; Weiss, P.; Beck, L.; et al. Phosphate-Dependent Regulation of MGP in Osteoblasts: Role of ERK1/2 and Fra-1. J. Bone Miner. Res. 2009, 24, 1856–1868. [Google Scholar] [CrossRef] [PubMed]
  182. Khoshniat, S.; Bourgine, A.; Julien, M.; Petit, M.; Pilet, P.; Rouillon, T.; Masson, M.; Gatius, M.; Weiss, P.; Guicheux, J.; et al. Phosphate-dependent stimulation of MGP and OPN expression in osteoblasts via the ERK1/2 pathway is modulated by calcium. Bone 2011, 48, 894–902. [Google Scholar] [CrossRef] [PubMed]
  183. Bozec, A.; Bakiri, L.; Jimenez, M.; Schinke, T.; Amling, M.; Wagner, E.F. Fra-2/AP-1 controls bone formation by regulating osteoblast differentiation and collagen production. J. Cell Biol. 2010, 190, 1093–1106. [Google Scholar] [CrossRef] [Green Version]
  184. Yates, A.J.; Oreffo, R.O.C.; Mayor, K.; Mundy, G.R. Inhibition of bone resorption by inorganic phosphate is mediated by both reduced osteoclast formation and decreased activity of mature osteoclasts. J. Bone Miner. Res. 1991, 6, 473–478. [Google Scholar] [CrossRef]
  185. O’Neill, E.; Awale, G.; Daneshmandi, L.; Umerah, O.; Lo, K.W.H. The roles of ions on bone regeneration. Drug Discov. Today 2018, 23, 879–890. [Google Scholar] [CrossRef]
  186. Hartwig, A. Role of magnesium in genomic stability. Mutat. Res./Fundam. Mol. Mech. Mutagen. 2001, 475, 113–121. [Google Scholar] [CrossRef]
  187. Sun, J.; Sun, Q.; Lu, S. From selenoprotein to endochondral ossification: A novel mechanism with microRNAs potential in bone related diseases? Med. Hypotheses 2011, 77, 807–811. [Google Scholar] [CrossRef]
  188. Sripanyakorn, S.; Jugdaohsingh, R.; Thompson, R.P.H.; Powell, J.J. Dietary silicon and bone health. Nutr. Bull. 2005, 30, 222–230. [Google Scholar] [CrossRef]
  189. Carlisle, E.M. Silicon: A requirement in bone formation independent of vitamin D1. Calcif. Tissue Int. 1981, 33, 27–34. [Google Scholar] [CrossRef]
  190. Jugdaohsingh, R.; Tucker, K.L.; Qiao, N.; Cupples, L.A.; Kiel, D.P.; Powell, J.J. Dietary silicon intake is positively associated with bone mineral density in men and premenopausal women of the Framingham Offspring cohort. J. Bone Miner. Res. 2004, 19, 297–307. [Google Scholar] [CrossRef] [Green Version]
  191. Nieves, J.W. Skeletal effects of nutrients and nutraceuticals, beyond calcium and vitamin D. Osteoporos. Int. 2013, 24, 771–786. [Google Scholar] [CrossRef]
  192. Hott, M.; de Pollak, C.; Modrowski, D.; Marie, P.J. Short-term effects of organic silicon on trabecular bone in mature ovariectomized rats. Calcif. Tissue Int. 1993, 53, 174–179. [Google Scholar] [CrossRef]
  193. Marie, P.J.; Ammann, P.; Boivin, G.; Rey, C. Mechanisms of action and therapeutic potential of strontium in bone. Calcif. Tissue Int. 2001, 69, 121–129. [Google Scholar] [CrossRef]
  194. Buehler, J.; Chappuis, P.; Saffar, J.L.; Tsouderos, Y.; Vignery, A. Strontium ranelate inhibits bone resorption while maintaining bone formation in alveolar bone in monkeys (Macaca fascicularis). Bone 2001, 29, 176–179. [Google Scholar] [CrossRef]
  195. Reginster, J.Y.; Kaufman, J.M.; Goemaere, S.; Devogelaer, J.P.; Benhamou, C.L.; Felsenberg, D.; Diaz-Curiel, M.; Brandi, M.L.; Badurski, J.; Wark, J.; et al. Maintenance of antifracture efficacy over 10 years with strontium ranelate in postmenopausal osteoporosis. Osteoporos. Int. 2012, 23, 1115–1122. [Google Scholar] [CrossRef] [Green Version]
  196. Verberckmoes, S.C.; De Broe, M.E.; D’Haese, P.C. Dose-dependent effects of strontium on osteoblast function and mineralization. Kidney Int. 2003, 64, 534–543. [Google Scholar] [CrossRef] [Green Version]
  197. Osredkar, J.; Sustar, N. Copper and zinc, biological role and significance of copper/zinc imbalance. J. Clin. Toxicol. 2011, 3, 0495. [Google Scholar] [CrossRef] [Green Version]
  198. Fleury, C.; Petit, A.; Mwale, F.; Antoniou, J.; Zukor, D.J.; Tabrizian, M.; Huk, O.L. Effect of cobalt and chromium ions on human MG-63 osteoblasts in vitro: Morphology, cytotoxicity, and oxidative stress. Biomaterials 2006, 27, 3351–3360. [Google Scholar] [CrossRef]
  199. Hallab, N.J.; Vermes, C.; Messina, C.; Roebuck, K.A.; Glant, T.T.; Jacobs, J.J. Concentration- and composition-dependent effects of metal ions on human MG-63 osteoblasts. J. Biomed. Mater. Res. 2002, 60, 420–433. [Google Scholar] [CrossRef]
  200. Anissian, L.; Stark, A.; Dahlstrand, H.; Granberg, B.; Good, V.; Bucht, E. Cobalt ions influence proliferation and function of human osteoblast-like cells. Acta Orthop. Scand. 2002, 73, 369–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Queally, J.M.; Devitt, B.M.; Butler, J.S.; Malizia, A.P.; Murray, D.; Doran, P.P.; O’Byrne, J.M. Cobalt ions induce chemokine secretion in primary human osteoblasts. J. Orthop. Res. 2009, 27, 855–864. [Google Scholar] [CrossRef] [PubMed]
  202. Wu, C.; Zhou, Y.; Fan, W.; Han, P.; Chang, J.; Yuen, J.; Zhang, M.; Xiao, Y. Hypoxia-mimicking mesoporous bioactive glass scaffolds with controllable cobalt ion release for bone tissue engineering. Biomaterials 2012, 33, 2076–2085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Fan, W.; Crawford, R.; Xiao, Y. Enhancing in vivo vascularized bone formation by cobalt chloride-treated bone marrow stromal cells in a tissue engineered periosteum model. Biomaterials 2010, 31, 3580–3589. [Google Scholar] [CrossRef] [Green Version]
  204. Birgani, Z.T.; Gharraee, N.; Malhotra, A.; Van Blitterswijk, C.A.; Habibovic, P. Combinatorial incorporation of fluoride and cobalt ions into calcium phosphates to stimulate osteogenesis and angiogenesis. Biomed. Mater. 2016, 11, 015020. [Google Scholar] [CrossRef]
  205. Kargozar, S.; Lotfibakhshaiesh, N.; Ai, J.; Mozafari, M.; Milan, P.B.; Hamzehlou, S.; Barati, M.; Baino, F.; Hill, R.G.; Joghataei, M.T. Strontium-and cobalt-substituted bioactive glasses seeded with human umbilical cord perivascular cells to promote bone regeneration via enhanced osteogenic and angiogenic activities. Acta Biomater. 2017, 58, 502–514. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of relationships between three essential components (cells, organics, inorganics) in maintaining bone homeostasis. Cell-derived organic molecules, such as growth factors and enzymes, modulate cellular activities; Osteoclasts release ions from the bone matrix during bone resorption, and in turn, ions act as molecular modulators of cellular activities and as components of apatite crystals being deposited into the bone matrix with the modulation of cells; Ions are co-factors to many enzymes, and ions are immobilized as apatite crystals into collagen fibrils from the bone structure at the nanoscale.
Figure 1. Schematic illustration of relationships between three essential components (cells, organics, inorganics) in maintaining bone homeostasis. Cell-derived organic molecules, such as growth factors and enzymes, modulate cellular activities; Osteoclasts release ions from the bone matrix during bone resorption, and in turn, ions act as molecular modulators of cellular activities and as components of apatite crystals being deposited into the bone matrix with the modulation of cells; Ions are co-factors to many enzymes, and ions are immobilized as apatite crystals into collagen fibrils from the bone structure at the nanoscale.
Jfb 14 00056 g001
Figure 2. A schematic illustration of the localized microenvironment at the bone remodeling site. Bone homeostasis is maintained by the balance between bone formation by osteoblasts and bone resorption by osteoclasts. During the bone remodeling process, organic molecules, such as enzymes, growth factors, and hormones, are released into the localized microenvironment, together with a mixture of inorganic components.
Figure 2. A schematic illustration of the localized microenvironment at the bone remodeling site. Bone homeostasis is maintained by the balance between bone formation by osteoblasts and bone resorption by osteoclasts. During the bone remodeling process, organic molecules, such as enzymes, growth factors, and hormones, are released into the localized microenvironment, together with a mixture of inorganic components.
Jfb 14 00056 g002
Figure 3. A schematic representation of the top view of unit cells of (a) stoichiometric hydroxyapatite; and (b) biological apatite crystals. Hydroxyl ions (OH) are positioned on the screw axes at every one-half of the unit cell, paralleling the c-axis. Calcium ions (Ca2+) are interspersed among tetrahedral phosphate ions (PO43−), and the marginal ones are shared with neighbor unit cells.
Figure 3. A schematic representation of the top view of unit cells of (a) stoichiometric hydroxyapatite; and (b) biological apatite crystals. Hydroxyl ions (OH) are positioned on the screw axes at every one-half of the unit cell, paralleling the c-axis. Calcium ions (Ca2+) are interspersed among tetrahedral phosphate ions (PO43−), and the marginal ones are shared with neighbor unit cells.
Jfb 14 00056 g003
Figure 4. A schematic illustration of the dissolution and precipitation process near the surface of an inorganic biomaterial in vivo in the ionic microenvironment created by cells and physiological fluid enriched by dissolved biomaterial.
Figure 4. A schematic illustration of the dissolution and precipitation process near the surface of an inorganic biomaterial in vivo in the ionic microenvironment created by cells and physiological fluid enriched by dissolved biomaterial.
Jfb 14 00056 g004
Figure 5. An illustration of the current understanding of the influence of ions on MSCs, osteoblasts, and osteoclasts.
Figure 5. An illustration of the current understanding of the influence of ions on MSCs, osteoblasts, and osteoclasts.
Jfb 14 00056 g005
Table 1. Summary of major biological influences of bone homeostasis-related bioactive inorganics at physiological and cellular levels.
Table 1. Summary of major biological influences of bone homeostasis-related bioactive inorganics at physiological and cellular levels.
IonRelated Disorders or DiseasesEffects on Cellular ActivitiesReferences
+
CaDeficiency: rickets, osteomalacia, and osteoporosis;
Overload: poor bone health, kidney stone formation, and abnormal heart and brain function
MSC mineralization, osteoblast cell proliferation, survival and differentiation, osteoclast cell apoptosisOsteoblast cell apoptosis, bone resorption[9,74,76,77,78,79,80]
PiDeficiency: impaired bone mineralization, dysfunction in blood, muscle, central nervous system, cardio and respiratory system;
Overload: kidney disease, cardiovascular disease, cancer, and skeletal disorder
Osteoblast and osteoclast cell apoptosis (high Pi level), osteoblastic differentiation and mineralization, bone resorption (low Pi level)Bone resorption (at high Pi levels)[50,75,81,82,83,84,85,86]
BDeficiency: reduced osteogenesis, inhibited bone formation, decreased bone volume, and reduced mechanical strengthMSC and osteoblast osteogenic differentiation and mineralization*[49,87,88,89,90,91,92]
CuDeficiency: abnormal bone formation with impaired quality and strength, severe neurological issues, or liver diseasesangiogenesis, innate antibacterial property, extracellular matrix formation*[4,93,94,95,96,97,98,99]
Ga*Bone formation and mineralizationOsteoclast differentiation, bone resorption[53,100,101,102]
MgDeficiency: impaired bone growth, disrupted mineral metabolism, and osteoporosisMSC osteogenic differentiation and mineralizationOsteoblast differentiation (high Mg level)[103,104,105,106,107,108,109,110,111,112]
FeDeficiency: overall loss in bone mass and density, impaired biomechanical strength Overload: metabolic bone diseases such as osteoporosis, altered bone microarchitecture, and reduced biomechanical strengthBone resorption (high Fe level)Osteoblast cell maturation and differentiation (high Fe level)[113,114,115,116]
MnDeficiency: abnormal bone growth, such as stunted bone growth and osteoporosis;
Overload: impaired bone development and neurotoxicity
Osteoblast proliferation, adhesion, and spreading, osteoblastic differentiation, collagen deposition, angiogenesis, and bone healing*[21,117,118,119,120,121,122,123]
SeDeficiency: impaired bone and cartilage metabolism, osteopenia, osteoporosis, and Kashin-Beck disease (together with iodine);
Overload: decreased mineral content, altered bone structure, and reduced biomechanical strength
***[74,124,125,126,127,128,129,130,131]
SiDeficiency: abnormal bone growthOsteoblast cell growth, proliferation, and differentiationOsteoclast formation, recruitment, and bone resorption, as well as osteoblast-induced osteoclastogenesis[132,133,134,135,136,137,138,139,140]
Sr*Pre-osteoblast cell replication and collagen synthesis, osteoblast cell proliferation, survival, differentiation, mineralization, osteoclast cell apoptosisOsteoclast cell survival, differentiation, osteoblast-induced osteoclastogenesis, and bone resorption[48,78,141,142,143,144,145,146,147,148,149,150,151,152,153]
ZnDeficiency: abnormal immune response, impaired wound healing, overall bone mass, and health, and bone turnover rateMSC viability, osteoblastic differentiation, and mineralization, osteoblast cell proliferation, differentiation, and mineralizationOsteoclastogenesis and bone resorption[154,155,156,157,158,159,160,161,162,163]
+ Promotive effect, − Inhibitive effect, * Not applicable, ** Unclear.
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

Mu, Y.; Du, Z.; Xiao, L.; Gao, W.; Crawford, R.; Xiao, Y. The Localized Ionic Microenvironment in Bone Modelling/Remodelling: A Potential Guide for the Design of Biomaterials for Bone Tissue Engineering. J. Funct. Biomater. 2023, 14, 56. https://doi.org/10.3390/jfb14020056

AMA Style

Mu Y, Du Z, Xiao L, Gao W, Crawford R, Xiao Y. The Localized Ionic Microenvironment in Bone Modelling/Remodelling: A Potential Guide for the Design of Biomaterials for Bone Tissue Engineering. Journal of Functional Biomaterials. 2023; 14(2):56. https://doi.org/10.3390/jfb14020056

Chicago/Turabian Style

Mu, Yuqing, Zhibin Du, Lan Xiao, Wendong Gao, Ross Crawford, and Yin Xiao. 2023. "The Localized Ionic Microenvironment in Bone Modelling/Remodelling: A Potential Guide for the Design of Biomaterials for Bone Tissue Engineering" Journal of Functional Biomaterials 14, no. 2: 56. https://doi.org/10.3390/jfb14020056

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