The Assessment of Bioactivity and Biological Responsiveness in Bioactive Glasses and Ceramics: A Review of Available Techniques
Abstract
1. Introduction
- (i)
- Metals are used in load-bearing applications due to their excellent mechanical strength and durability. However, once implanted in the body, they may be susceptible to corrosion;
- (ii)
- Polymers, both natural and synthetic, offer good versatility in terms of degradation rates and processing, making them suitable for soft tissue engineering and drug delivery systems;
- (iii)
- Composites, composed of two or more materials, are engineered to combine the complementary properties of each phase.
- (a)
- Nearly inert ceramics, which exhibit minimal interaction with biological tissues and are commonly used in joint prostheses and cardiovascular devices;
- (b)
- Surface-reactive ceramics, which actively interact with biological fluids to enhance integration with surrounding tissues;
- (c)
- esorbable ceramics, which, as the name suggests, gradually degrade and are absorbed by the body.
- (i)
- Tricalcium phosphate (TCP) [12], a calcium salt of phosphoric acid (Ca3(PO4)2), which exists in three polymorphic forms: α-TCP (stable between ~1120 °C and 1465 °C), α′-TCP (stable at temperatures > 1430 °C), and β-TCP (stable at room temperature and transforming into α-TCP at ~1120 °C) [12]. From a biological perspective, both α-TCP and β-TCP are biodegradable and bioactive, with a resorption rate higher than crystalline apatite [13];
- (ii)
- Synthesized apatite, a ceramic similar in composition to bone mineral. The term apatite refers to a group of crystalline compounds including HAp (Ca10(PO4)6(OH)2), fluorapatite, and the composite fluoro-hydroxyapatite (FHA) [14]. Synthetic HAp has been widely used in biomedical applications, including porous implants, powders, and coatings for metallic prostheses to enhance bioactive fixation [14]. The incorporation of HAp coatings has been shown to promote bone growth along the implant surface, forming a mechanically strong interface. Additionally, HAp particles can be integrated into polymer matrices such as polyethylene, forming bioactive composites that play a crucial role in bone repair and replacement, particularly in the reconstruction of the middle ear;
- (iii)
- Apatite-wollastonite glass-ceramics (A-W) [15], a bioceramic that creates a bone-like apatite layer on its surface in vivo, enabling strong bonding with surrounding tissues. The A/W glass-ceramic was studied by Prof. Kokubo and colleagues in 1980s and demonstrated excellent bioactivity and mechanical properties, making it a material with great potential for mechanically resilient biomedical applications [16]. Moreover, its composition and microstructure enhance long-term stability in physiological environments [17].
2. In Vitro Static and Dynamic Approaches for Evaluating Bioactivity and Biological Responsiveness
- -
- Low flow rates (~0.04 mL/min) correspond to high ion concentrations in the solution and a low dissolution rate [47].
- -
- With high flow rates (~0.6 mL/min), dissolution occurs uniformly and is influenced by surface reactions [47].
3. In Vitro Assays for the Study of Bioactivity
3.1. Simulated Body Fluid (SBF)
- (i)
- Kim et al. [66] demonstrated that the HAp layer formed in SBF differs from bone apatite in both composition and structure, attributing these differences to SBF’s higher Cl− content and lower HCO3− concentration compared to blood plasma. Based on these findings, Oyane et al. [67] revised the original SBF and introduced new variants (c-SBF, r-SBF, i-SBF, and m-SBF) with ion concentrations more closely matching those of blood plasma (Table 3) and supersaturated with respect to HAp. The results demonstrated that upon storage, r-SBF and i-SBF showed no changes in ion concentrations but exhibited a decrease in HCO3− and an increase in pH. In contrast, c-SBF and m-SBF maintained stable ion concentrations for a longer period, indicating that r-SBF and i-SBF are less stable in terms of ionic composition. Among these formulations, m-SBF was found to have the most optimal ion composition for assessing bioactivity in vitro and for the biomimetic synthesis of HAp.
- (ii)
- Müller et al. [68] explored the role of HCO3− by proposing an SBF formulation with varying bicarbonate concentrations, as these ions affect the chemical composition and the structure of the calcium phosphate that form. At low HCO3− concentrations, only carbonate hydroxyapatite (HCA), in which carbonate ions replace phosphate groups, is formed. Conversely, at higher bicarbonate concentrations, HCA crystals in which carbonate ions substitute hydroxyl groups are synthesized.
- (iii)
- Mei et al. [69] investigated the influence of proteins on the reaction kinetics of BGs in modified SBF solutions. In their study, proteins such as bovine serum albumin were added to better mimic the biological environment. The presence of proteins affected the surface reactions of BGs in vitro, modifying the dissolution rate as well as the composition and structural organization of the layer formed on the material’s surface.
- (i)
- presented several examples that contradict this assumption, including BGs, β-TCP, HAp, dicalcium phosphate dihydrate (DCPD), and calcium sulfate hemihydrate (CSH). Discrepancies in SBF testing arise from material solubility and local changes in supersaturation. Highly reactive BGs increase local pH, reducing HAp solubility and accelerating apatite nucleation, potentially overestimating in vitro bioactivity. Similarly, DCPD or CSH release high amounts of calcium and phosphate ions, inducing rapid HAp precipitation. However, they dissolve too quickly in vivo to establish a stable bond with bone, resulting in false positives.
- (ii)
- showed that the selection of an SBF solution is often arbitrary and proposed an iterative method to refine the in vitro approach. This involves selecting a simple SBF that mimics blood serum properties, testing reference materials, and investigating the influence of additives to improve the correlation between in vitro and in vivo results, reducing the risk of false positives or false negatives.
- (i)
- Zhang et al. [75] investigated the effect of fluid circulation on the in vitro bioactivity of 45S5 Bioglass® granules (500–800 μm), demonstrating that dynamic conditions significantly influence HAp formation. Under static conditions, reaction layers were thicker, often presenting a distinct calcium-phosphate coating on the top of the silica gel layer. Conversely, under dynamic conditions, the reaction deposits were thinner and more homogeneous, with calcium-phosphate inclusions appearing as a single mixed layer. HAp formation under static conditions depended on the position of the samples, whereas under dynamic conditions, a uniform layer formed on all particles.
- (ii)
- Similarly, Ràmila et al. [76] conducted a comparative study on the in vitro bioactivity of a sol–gel-derived SiO2-CaO-P2O5 glass, evaluating its behavior in static and dynamic SBF conditions. Under dynamic conditions, Ca2+ concentration and pH remained stable and comparable to those of human blood plasma, making this approach more suitable for predicting in vivo bioactivity.
- (iii)
- Following the same approach, Kang et al. [77] examined the in vitro bioactivity of a poly(L-lactic acid)/β-TCP porous scaffold. Their results show that fluid circulation significantly reduced the degradation rate of molecular weight and compressive strength.
3.2. Tris-Buffer Solution (TRIS)
3.3. Phosphate-Buffer Saline (PBS) Solution
- (i)
- Yusof et al. [55] explored the impact of incorporating B2O3 into 45S5 Bioglass®, emphasizing the importance of understanding element dissolution from BGs in PBS, as this correlates with the material’s bone-bonding ability in vivo. Their study proposed an Equation (1), based on prior research [98], to determine the dissolution rate of each element from the sample immersed in the physiological solution [55]:
- (ii)
- Zhou et al. [99] investigated the in vitro degradation behavior of BG-polymer composites in PBS, demonstrating that the incorporation of BG particles altered the dissolution kinetics of the polymer. The acidic degradation products of the polymeric matrix interacted with PBS and were neutralized by the alkaline degradation products released from the BG particles.
- (iii)
- Loh et al. [97] studied calcium-based 45S5 BG pellets in PBS, reporting the formation of carbonate apatite, fluorapatite, and calcite phases. Their study highlighted that the interaction between BGs and PBS accelerates over time, leading to progressive HAp development on the material’s surface.
- (a)
- Most studies have been conducted with static immersion in PBS, while dynamic testing in this medium has not yet been investigated in the literature; dynamic approaches are more commonly reported with other buffered solutions, such as SBF or TRIS;
- (b)
- Cases of false positives or false negatives in PBS have received limited attention in the literature, even though Bohner et al. [73] conducted similar tests as those reported for SBF, and their conclusions were consistent with previous findings.
3.4. Simulated Wound Fluid (SWF)
3.5. Comparative Analysis of Simulated Physiological Solutions for In Vitro Bioactivity Assessment
4. Cell Culture Assays for the Study of Biological Responsiveness
- (i)
- Cytotoxicity tests [54]: Used as a preliminary method to assess the safety of a biomaterial. These tests evaluate cell lysis (cell death), inhibition of cell growth, and the ability of cells to survive and proliferate after exposure to potentially toxic materials or substances (e.g., colony formation assays). Additional cellular effects induced by the biomaterial may also be considered. ISO 10993-5 recommends three methods for cytotoxicity testing: extract dilution, direct contact, and indirect contact methods. These are described in detail in the following paragraphs.
- (ii)
- Hemocompatibility tests [108]: Used to evaluate the effects of biomaterials when in contact with blood. A common example is haemolysis test, which determines the extent of red blood cell lysis and the release of hemoglobin induced by the biomaterials.
- (iii)
- Genotoxicity tests [107]: Used to detect gene mutations and changes in chromosome structure or number caused by biomaterials.
- (iv)
- Sensitization test: Used to estimate the potential of a biomaterial to cause contact sensitization.
- (v)
- Irritation test: Used to evaluate the irritation potential of materials when applied to a specific site. The tests are conducted in accordance with ISO 10993-1 and ISO 10993-2.
- (vi)
- Scratch test [109]: A simple and cost-effective method for measuring cell migration. It involves creating a “scratch” in a cell monolayer and capturing microscopic images immediately afterward and at regular intervals during incubation to monitor cell movement. Compared to other cell culture assays, the scratch test typically requires a longer duration and a larger quantity of cells and reagents.
5. Methods for In Vitro Biological Cellular Assays
6. Cell Type Selection for In Vitro Biological Evaluation
- (i)
- For cytotoxicity tests, the standardized protocol ISO 10993-5 [111] recommends the use of: (a) L-929 fibroblast cells for MTT and XTT assays, due to their high sensitivity to toxic substances and reproducible growth characteristics; (b) BALB3T3 fibroblasts for Neutral Red (NR) assay, because of their rapid proliferation, which ensures consistent and comparable results. Although not formally included in ISO 10993-5, the Trypan Blue exclusion assay is frequently used with L-929 fibroblasts, which are specified in the standard protocol.
- (ii)
- For proliferation assays (e.g., BrdU or EdU assays), human mesenchymal stem cells (hMSCs) and pre-osteoblasts such as MC3T3-E1 are commonly used due to their relevance in bone regeneration studies.
- (iii)
- In osteogenic differentiation assays, including alkaline phosphatase activity (ALP) test and Alizarin red staining, hMSCs are widely used because of their multipotency, while MC3T3-E1 and Saos-2 osteoblast-like cells serve as established models for studying osteoblastic behavior.
- (iv)
- Gene and protein expression studies (e.g., qPCR, immunocytochemistry) similarly rely on osteogenic, pre-osteoblast, or endothelial cell types to evaluate the molecular pathways involved in differentiation and mineralization.
- (v)
- For assays aimed at genotoxicity evaluation, such as the comet assay, fibroblast cell lines (e.g., L-929, C165) or primary cells (e.g., hMSCs) are used, depending on the specific objectives of the study.
7. Cytotoxicity and Viability Assays
7.1. MTT Assay
7.2. XTT Assay
- (i)
- Mitri et al. [130] evaluated the viability of MSCs exposed to extracts from dense and porous HAp/β-TCP ceramic granules using a multi-assay approach combining XTT, Neutral Red, and Crystal Violet tests. The authors found that ceramic extracts significantly enhanced mitochondrial dehydrogenase activity without inducing cytotoxicity. However, in earlier studies, Mitri et al. reported cytotoxic effects from porous ceramic extracts, which were attributed to the possible release of microparticles during sintering. The observed loss in cell viability was associated with growth inhibition caused by BGs when cells were cultured in the presence of varying sample concentrations.
- (ii)
- Deliormanli [131] used the XTT assay to evaluate the osteoblast cell response to silicate-based 13–93 BG fibers (54.6% SiO2, 22.1% CaO, 7.9% K2O, 7.7% MgO, 6.0% Na2O, and 1.7% P2O5) fabricated via sol–gel processing and electrospinning. The assay revealed no cytotoxicity of the scaffolds but indicated that cells may penetrate the porous structure of the biomaterial. This indicates that spectrophotometric quantification may be influenced by cell infiltration into the porous structure of the tested material, potentially leading to false negative results or underestimation of cell viability.
7.3. Alamar Blue® Assay
7.4. Neutral Red Uptake (NR) Assay
7.5. Trypan Blue Exclusion Test
7.6. Crystal Violet Staining
8. Proliferation Assay
8.1. Bromodeoxyuridine (BrdU) Assay
- (a)
- The study by Bielby et al. [159] highlights this critical limitation. The authors used BrdU to assess the proliferation of primary murine and human osteoblasts in response to the dissolution products of 58S BG (60% SiO2, 36% CaO, 4% P2O5). Murine osteoblasts showed a 100% increase in BrdU uptake by day 6, indicating a strong mitogenic response. In contrast, human osteoblasts exhibited an early inhibition of BrdU incorporation (day 2), followed by stimulation (day 4), and then inhibition again (day 6). These findings suggest that the soluble ions released from the glass can modulate cell proliferation, but the response is highly cell-type dependent. The inhibitory effects observed at higher glass concentrations were likely due to increased medium pH and cytotoxicity from elevated cation levels, which were not present at lower concentrations.
- (b)
- While Bielby et al. [159], reported a dose-dependent inhibition of BrdU incorporation at high concentrations of undoped 58S BG, other studies suggest that modifying the glass composition can mitigate these adverse effects. For instance, Malavasi et al. [154] investigated Ce-doped BGs based on the 45S5 composition using long-bone osteocyte-like (MLO-Y4) and fibroblast (NIH/3T3) cell lines, and observed no cytotoxicity or inhibition of proliferation, even at elevated BG concentrations. On the contrary, BrdU incorporation increased with the cerium content in the glasses.
8.2. 5-Ethynyl-2′-deoxyuridine (EdU) Assay
9. Differentiation Assays
9.1. Alizarin Red Staining
- (i)
- Zhao et al. [167] employed the Alizarin Red staining technique to evaluate the mineralization potential of bone marrow stromal cells (BMSCs) cultured with extracts from 45S5, β-TCP, and a phosphate-rich bioactive glass containing no Na2O (PSC). Their results showed that PSC significantly enhanced calcium deposition compared to the other materials, as evidenced by more intense Alizarin Red staining and higher absorbance values. The relatively stable pH environment resulting from the release of silicate, calcium, and phosphate ions from PSC supported improved cell viability and mineralization and contributed to the enhanced proliferation of BMSCs.
- (ii)
- The results reported by Vargas et al. [169] support the findings of Zhao et al. [167] regarding both the effectiveness of Alizarin Red staining and the influence of released ions on mineralization. Additionally, Vargas et al. proposed a dual-staining method combining Alizarin Red with Alcian blue—a polyvalent basic dye—for a more comprehensive evaluation of the biological responsiveness [169]. This approach may help mitigate the risk of false-positive results. Alizarin Red may also bind non-specifically to calcium-containing residues [168], such as precipitated calcium phosphate derived from the culture medium or degradation by-products of BG. This non-specific binding may lead to an overestimation of osteogenic differentiation. The inclusion of Alcian Blue, which stains sulfated glycosaminoglycans, aids in distinguishing genuine mineralization from such artifacts.
9.2. Alkaline Phosphatase (ALP) Activity Assay
10. Gene and Protein Expression Assays
10.1. Quantitative Polymerase Chain Reaction (qPCR)
- (i)
- Mosaddada et al. [166] used qPCR to analyze the osteogenic differentiation of cells seeded on collagen/Sr-doped BG scaffolds. Increasing Sr content led to an upregulation of gene and protein expression levels, which correlated with enhanced mineralization and bone regeneration.
- (ii)
- Zhang et al. [181] employed real-time qPCR (RT-qPCR) to investigate the osteogenic potential of BG-ceramic (BGC) coatings. PCR analysis was performed on three experimental samples, with results reported as target gene expression levels. These findings confirmed a dose-dependent significant upregulation of key osteogenic genes, including ALP, osteocalcin, and Runx2, in bone marrow MSCs cultured on BG-coatings. Higher BG content induced stronger gene expression responses.
- (iii)
- Furthermore, Morales-Hernandez et al. [177] applied qPCR to assess osteogenic gene expression in normal human osteoblasts (NHOsts) cultured on macroporous composite scaffolds composed of PLG and different bioceramics (HAp, TCP, and 45S5 Bioglass®). Their results demonstrated that BG-containing scaffolds significantly up-regulated the expression of bone matrix proteins, such as COL1A1 and SPARC, compared to HAp- and TCP-based composites, supporting the enhanced osteogenic potential induced by BG. BGs degrade more rapidly than HAp and TCP, triggering cellular processes through the release of ionic degradation products, such as silica ions. Overall, these observations also reinforce the concept that composite systems with BGs or bioceramics represent a highly promising class of materials, which can be engineered to enhance biological responsiveness [182,183].
10.2. Immunocytochemistry
- (i)
- Trandas et al. [185], who investigated Eu- and Ag-doped BG thin films. Using immunocytochemistry, they visualized cytoskeletal organization—particularly actin filaments—in pre-osteoblasts.
- (ii)
- Similarly, Lavric et al. [186] compared BG thin films doped with Ag and Sm, produced via pulse laser deposition (PLD) and spin coating (SC). Cell adhesion and viability were evaluated using immunocytochemistry with mouse pre-osteoblasts. Their in vitro results showed that the samples supported mineralization and promoted cell adhesion and proliferation, attributed to the presence of dopant ions.
11. Genotoxicity Assay
Comet Assay
12. Comparison Between In Vitro Biological Responsiveness Tests
13. Conclusions
- -
- The use of revised SBF (r-SBF) with lower calcium and magnesium concentrations;
- -
- Preventing an excessive precipitation, ensuring an adequate volume of SBF;
- -
- Incorporating organic components to create a more realistic environment.
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
Abbreviations
BG | Bioactive glass |
TCP | Tri-calcium phosphate |
A-W | Apatite-wollastonite |
SBF | Simulated body fluid |
TRIS | Tris-buffer |
PBS | Phosphate-buffer saline solution |
SWF | Simulated wound fluid |
RM | Regenerative medicine |
HAp | Hydroxyapatite |
HCA | Carbonate hydroxyapatite |
FHA | Fluor-hydroxyapatite |
TE | Tissue engineering |
MQ | Melt-quench technique |
SG | Sol–gel process |
XRD | X-Ray diffraction spectroscopy |
FT-IR | Fourier-transform infrared spectroscopy |
SEM | Scanning electron microscopy |
OOC | Organ-on-a-chip |
DCPD | Dicalcium phosphate dihydrate |
CSH | Calcium sulfate hemihydrate |
MTT | Methyl Thyazolyl Tetrazolium |
NR | Neutral Red uptake assay |
BrdU | Bromodeoxyuridine assay |
EdU | 5-ethynyl-2′-deoxyuridine assay |
ALP | Alkaline phosphatase activity |
qPCR | Quantitative Polymerase Chain Reaction |
References
- Ratner, B.D.; Bryant, S.J. Biomaterials: Where we have been and where we are going. Annu. Rev. Biomed. Eng. 2004, 6, 41–75. [Google Scholar] [CrossRef]
- Hench, L.L. Bioceramics: From Concept to Clinic. J. Am. Ceram. Soc. 1991, 74, 1487–1510. [Google Scholar] [CrossRef]
- Martelli, A.; Bellucci, D.; Cannillo, V. Additive Manufacturing of Polymer/Bioactive Glass Scaffolds for Regenerative Medicine: A Review. Polymers 2023, 15, 2473. [Google Scholar] [CrossRef]
- Mason, C.; Dunnill, P. A Brief Definition of Regenerative Medicine. Regen. Med. 2008, 3, 1–5. [Google Scholar] [CrossRef] [PubMed]
- Williams, D.F. The Williams Dictionary of Biomaterials; Liverpool University Press: Liverpool, UK, 1999. [Google Scholar]
- Jablonská, E.; Horkavcová, D.; Rohanová, D.; Brauer, D.S. A review of: In vitro cell culture testing methods for bioactive glasses and other biomaterials for hard tissue regeneration. J. Mater. Chem. B 2020, 8, 10941–10953. [Google Scholar] [CrossRef]
- Mahdi, N.M.S.; Hassan, A.K.; Al-Hasani, F.J.; Al-Jawher, W.A.M. Classification of Biomaterials and Their Applications. J. Port. Sci. Res. 2024, 7, 281–299. [Google Scholar] [CrossRef]
- Vallet-Regí, M. Ceramics for medical applications. J. Chem. Soc. Dalton Trans. 2001, 2, 97–108. [Google Scholar] [CrossRef]
- Dorozhkin, S.V. Current state of bioceramics. J. Ceram. Sci. Technol. 2018, 9, 353–370. [Google Scholar] [CrossRef]
- 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]
- Hulbert, S.F.; Hench, L.L.; Forbers, D.; Bowman, L.S. History of Bioceramics. Ceram. Int. 1982, 8, 131–140. [Google Scholar] [CrossRef]
- Carrodeguas, R.G.; De Aza, S. Tricalcium phosphate: Synthesis, properties and biomedical applications. Acta Biomater. 2011, 7, 3536–3546. [Google Scholar] [CrossRef]
- Lowe, B.; Ottensmeyer, M.P.; Xu, C.; He, Y.; Ye, Q.; Troulis, M.J. The Regenerative Applicability of Bioactive Glass and Beta-Tricalcium Phosphate in Bone Tissue Engineering: A Transformation Perspective. J. Funct. Biomater. 2019, 10, 16. [Google Scholar] [CrossRef]
- Jantová, S.; Theiszová, M.; Letašiová, S.; Birošová, L.; Palou, T.M. In vitro effects of fluor-hydroxyapatite, fluorapatite and hydroxyapatite on colony formation, DNA damage and mutagenicity. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2008, 265, 139–144. [Google Scholar] [CrossRef]
- Dyson, J.A.; Genever, P.G.; Dalgarno, K.W.; Wood, D.J. Development of custom-built bone scaffolds using mesenchymal stem cells and apatite-wollastonite glass-ceramics. Tissue Eng. 2007, 13, 2891–2901. [Google Scholar] [CrossRef]
- Kokubo, T.; Ito, S.; Huang, Z.T.; Hayashi, T.; Sakka, S.; Kitsugi, T.; Yamamuro, T. P-rich layer formed on high-strength bioactive glass-ceramic A-W. J. Biomed. Mater. Res. 1990, 24, 331–343. [Google Scholar] [CrossRef] [PubMed]
- Cannillo, V.; Pierli, F.; Sampath, S.; Siligardi, C. Thermal and physical characterisation of apatite/wollastonite bioactive glass–ceramics. J. Eur. Ceram. Soc. 2009, 29, 611–619. [Google Scholar] [CrossRef]
- Hench, L.L.; Polak, J.M. Third-Generation biomedical materials. Science 2002, 295, 1014–1017. [Google Scholar] [CrossRef]
- Bellucci, D.; Mazzilli, A.; Martelli, A.; Mecca, F.G.; Bonacorsi, S.; Lofaro, F.D.; Boraldi, F.; Quaglino, D.; Cannillo, V. Enrichment of strontium and magnesium improves the physical, mechanical and biological properties of bioactive glasses undergoing thermal treatments: New cues for biomedical applications. Ceram. Int. 2024, 50, 52819–52837. [Google Scholar] [CrossRef]
- Mecca, F.G.; Bellucci, D.; Cannillo, V. Effect of Thermal Treatments and Ion Substitution on Sintering and Crystallization of Bioactive Glasses: A Review. Materials 2023, 16, 4651. [Google Scholar] [CrossRef]
- Kaur, G.; Pandey, O.P.; Singh, K.; Homa, D.; Scott, B.; Pickrell, G. A review of bioactive glasses: Their structure, properties, fabrication and apatite formation. J. Biomed. Mater. Res. A 2014, 102, 254–274. [Google Scholar] [CrossRef] [PubMed]
- Brauer, D.S. Bioactive glasses—Structure and properties. Angew. Chem. Int. Ed. 2015, 54, 4160–4181. [Google Scholar] [CrossRef]
- Hench, L.L.; Jones, J.R. Bioactive glasses: Frontiers and Challenges. Front. Bioeng. Biotechnol. 2015, 3, 194. [Google Scholar] [CrossRef]
- Jones, J.R. Review of bioactive glass: From Hench to hybrids. Acta Biomater. 2013, 9, 4457–4486. [Google Scholar] [CrossRef] [PubMed]
- Baino, F.; Hamzehlou, S.; Kargozar, S. Bioactive glasses: Where are we and where are we going? J. Funct. Biomater. 2018, 9, 25. [Google Scholar] [CrossRef] [PubMed]
- Hench, L.L. The story of Bioglass®. J. Mater. Sci. Mater. Med. 2006, 17, 967–978. [Google Scholar] [CrossRef]
- 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]
- Pedone, A.; Charpentier, T.; Malavasi, G.; Menziani, M.C. New insights into the atomic structure of 45S5 bioglass by means of solid-state NMR spectroscopy and accurate first-principles simulations. Chem. Mater. 2010, 22, 5644–5652. [Google Scholar] [CrossRef]
- Andersson, O.H.; Liu, G.; Karlsson, K.H.; Niemi, L.; Miettinen, J.; Juhanoja, J. In vivo behaviour of glasses in the SiO2-Na2O-CaO-P2O5-Al2O3-B2O3 system. J. Mater. Sci. Mater. Med. 1990, 1, 219–227. [Google Scholar] [CrossRef]
- Hupa, L.; Lindfors, N.C. Bioactive Glass S 53 P 4—From a Statistically Suggested Composition to Clinical Success. In Bioactive Glasses and Glass-Ceramics: Fundamentals and Applications; Baino, F., Kargozar, S., Eds.; John Wiley & Sons, Inc.: Hoboke, NJ, USA, 2022; pp. 33–59. [Google Scholar] [CrossRef]
- Sergi, R.; Bellucci, D.; Cannillo, V. A comprehensive review of bioactive glass coatings: State of the art, challenges and future perspectives. Coatings 2020, 10, 757. [Google Scholar] [CrossRef]
- Delpino, G.P.; Borges, R.; Zambanini, T.; Joca, J.F.S.; Gaubeur, I.; de Souza, A.C.S.; Marchi, J. Sol-gel-derived 58S bioactive glass containing holmium aiming brachytherapy applications: A dissolution, bioactivity, and cytotoxicity study. Mater. Sci. Eng. C 2021, 119, 111595. [Google Scholar] [CrossRef]
- Thomas, A.; Bera, J. Sol–gel synthesis and in vitro bioactivity of glass–ceramics in SiO2–CaO–Na2O–P2O5 system. J. Solgel. Sci. Technol. 2016, 80, 411–416. [Google Scholar] [CrossRef]
- Furlan, R.G.; Correr, W.R.; Russi, A.F.C.; da Costa Iemma, M.R.; Trovatti, E.; Pecoraro, É. Preparation and characterization of boron-based bioglass by sol−gel process. J. Solgel. Sci. Technol. 2018, 88, 181–191. [Google Scholar] [CrossRef]
- Munyensanga, P.; Dahdah, M.; Bricha, M.; Semlali, A.; El Mabrouk, K. In vitro assessment and surface morphology of copper-silver co-doped ordered mesoporous antifungal bioactive glasses. Ceram. Int. 2024, 50, 17154–17165. [Google Scholar] [CrossRef]
- Jones, J.R.; Hench, L.L. Factors Affecting the Structure and Properties of Bioactive Foam Scaffolds for Tissue Engineering. J. Biomed. Mater. Res. B Appl. Biomater. 2004, 68, 36–44. [Google Scholar] [CrossRef] [PubMed]
- Nawaz, A.; Bano, S.; Yasir, M.; Wadood, A.; Rehman, M.A.U. Ag and Mn-doped mesoporous bioactive glass nanoparticles incorporated into the chitosan/gelatin coatings deposited on PEEK/bioactive glass layers for favorable osteogenic differentiation and antibacterial activity. Mater. Adv. 2020, 1, 1273–1284. [Google Scholar] [CrossRef]
- Aqib, R.; Kiani, S.; Bano, S.; Wadood, A.; Rehman, M.A.U. Ag–Sr doped mesoporous bioactive glass nanoparticles loaded chitosan/gelatin coating for orthopedic implants. Int. J. Appl. Ceram. Technol. 2021, 18, 544–562. [Google Scholar] [CrossRef]
- Nawaz, Q.; Rehman, M.A.U.; Burkovski, A.; Schmidt, J.; Beltrán, A.M.; Shahid, A.; Alber, N.K.; Peukert, W.; Boccaccini, A.R. Synthesis and characterization of manganese containing mesoporous bioactive glass nanoparticles for biomedical applications. J. Mater. Sci. Mater. Med. 2018, 29, 64. [Google Scholar] [CrossRef]
- Mozafari, M.; Banijamali, S.; Baino, F.; Kargozar, S.; Hill, R.G. Calcium carbonate: Adored and ignored in bioactivity assessment. Acta Biomater. 2019, 91, 35–47. [Google Scholar] [CrossRef]
- Cannio, M.; Bellucci, D.; Roether, J.A.; Boccaccini, D.N.; Cannillo, V. Bioactive glass applications: A literature review of human clinical trials. Materials 2021, 14, 5440. [Google Scholar] [CrossRef] [PubMed]
- Shin, K.; Acri, T.; Geary, S.; Salem, A.K. Biomimetic Mineralization of Biomaterials Using Simulated Body Fluids for Bone Tissue Engineering and Regenerative Medicine. Tissue Eng. Part A 2017, 23, 1169–1180. [Google Scholar] [CrossRef]
- Xia, T.; Hamilton, R.F.; Bonner, J.C.; Crandall, E.D.; Elder, A.; Fazlollahi, F.; Girtsman, T.A.; Kim, K.; Mitra, S.; Ntim, S.A.; et al. Interlaboratory Evaluation of in Vitro Cytotoxicity and Inflammatory Responses to Engineered Nanomaterials: The NIEHS Nano GO Consortium. Environ. Health Perspect. 2013, 121, 683. [Google Scholar] [CrossRef]
- Park, S.Y.; Yi, S.M.; On, S.W.; Che, S.A.; Lee, J.Y.; Yang, B.E. Evaluation of low-crystallinity apatite as a novel synthetic bone graft material: In vivo and in vitro analysis. J. Dent. 2025, 154, 105597. [Google Scholar] [CrossRef]
- Lutišanová, G.; Kuzielová, E.; Palou, M.T.; Kozánková, J. Static and dynamic in vitro test of bioactivity of glass ceramics. Ceram.–Silikáty 2011, 55, 106–113. [Google Scholar]
- Rohanová, D.; Horkavcová, D.; Helebrant, A.; Boccaccini, A.R. Assessment of in vitro testing approaches for bioactive inorganic materials. J. Non-Cryst. Solids 2016, 432, 53–59. [Google Scholar] [CrossRef]
- Stiller, A.; Engblom, M.; Karlström, O.; Lindén, M.; Hupa, L. Impact of fluid flow rate on the dissolution behavior of bioactive glass S53P4. J. Non-Cryst. Solids 2023, 607, 122219. [Google Scholar] [CrossRef]
- Fagerlund, S.; Ek, P.; Hupa, L.; Hupa, M. Dissolution kinetics of a bioactive glass by continuous measurement. J. Am. Ceram. Soc. 2012, 95, 3130–3137. [Google Scholar] [CrossRef]
- Sinitsyna, P.; Engblom, M.; Hupa, L. Analysis and modelling of in vitro bioactivity for bioactive glass microspheres and granules in continuous fluid flow conditions. J. Non-Cryst. Solids 2024, 637, 123029. [Google Scholar] [CrossRef]
- Siekkinen, M.; Karlström, O.; Hupa, L. Dissolution of bioactive glass S53P4 in a three-reactor cascade in continuous flow conditions. Open Ceram. 2023, 13, 100327. [Google Scholar] [CrossRef]
- Siekkinen, M.; Karlström, O.; Hupa, L. Effect of local ion concentrations on the in vitro reactions of bioactive glass 45S5 particles. Int. J. Appl. Glass. Sci. 2022, 13, 695–707. [Google Scholar] [CrossRef]
- Syahruddin, M.H.; Anggraeni, R.; Ana, I.D.; Ana, I.D. A Microfluidic Organ-On-A-Chip: Into the Next Decade of Bone Tissue Engineering Applied in Dentistry. Future Sci. 2023, 9, FSO902. [Google Scholar] [CrossRef]
- Kokubo, T.; Takadama, H. How useful is SBF in predicting in vivo bone bioactivity? Biomaterials 2006, 27, 2907–2915. [Google Scholar] [CrossRef]
- Bellucci, D.; Salvatori, R.; Cannio, M.; Luginina, M.; Orru, R.; Montinaro, S.; Anesi, A.; Chiarini, L.; Cao, G.; Cannillo, V. Bioglass and bioceramic composites processed by Spark Plasma Sintering (SPS): Biological evaluation Versus SBF test. Biomed. Glas. 2018, 4, 21–31. [Google Scholar] [CrossRef]
- Yusof, N.N.; Aziz, S.M.; Noor, F.M.; Yaacob, S.N.S.; Hashim, S. A novel borate-based 45S5 Bioglass®: In vitro assessment in phosphate-buffered saline solution. J. Non-Cryst. Solids 2022, 596, 121843. [Google Scholar] [CrossRef]
- ISO 23317:2007; Implants for Surgery—In Vitro Evaluation for Apatite-Forming Ability of Implant Materials. Available online: https://www.iso.org/standard/41446.html (accessed on 15 September 2025).
- Maçon, A.L.B.; Kim, T.B.; Valliant, E.M.; Goetschius, K.; Brow, R.K.; Day, D.E.; Hoppe, A.; Boccaccini, A.R.; Kim, I.Y.; Ohtsuki, C.; et al. A unified in vitro evaluation for apatite-forming ability of bioactive glasses and their variants. J. Mater. Sci. Mater. Med. 2015, 26, 115. [Google Scholar] [CrossRef]
- Rohanová, D.; Boccaccini, A.R.; Horkavcová, D.; Bozděchová, P.; Bezdička, P.; Častorálová, M. Is non-buffered DMEM solution a suitable medium for in vitro bioactivity tests? J. Mater. Chem. B 2014, 2, 5068–5076. [Google Scholar] [CrossRef]
- Pirayesh, H.; Nychka, J.A. Sol-gel synthesis of bioactive glass-ceramic 45S5 and its in vitro dissolution and mineralization behavior. J. Am. Ceram. Soc. 2013, 96, 1643–1650. [Google Scholar] [CrossRef]
- Lopes, J.H.; Mazali, I.O.; Landers, R.; Bertran, C.A. Structural investigation of the surface of bioglass 45S5 enriched with calcium ions. J. Am. Ceram. Soc. 2013, 96, 1464–1469. [Google Scholar] [CrossRef]
- Rehman, I.; Hench, L.L.; Bonfield, W.; Smith, R. Analysis of surface layers on bioactive glasses. Biomaterials 1994, 15, 865–870. [Google Scholar] [CrossRef]
- Zheng, K.; Solodovnyk, A.; Li, W.; Goudouri, O.M.; Stähli, C.; Nazhat, S.N.; Boccaccini, A.R. Aging time and temperature effects on the structure and bioactivity of gel-derived 45S5 glass-ceramics. J. Am. Ceram. Soc. 2015, 98, 30–38. [Google Scholar] [CrossRef]
- Bano, S.; Akhtar, M.; Yasir, M.; Maqbool, M.S.; Niaz, A.; Wadood, A.; Rehman, M.A.U. Synthesis and characterization of silver-strontium (Ag-Sr)-doped mesoporous bioactive glass nanoparticles. Gels 2021, 7, 34. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Wang, H.; Zhao, S.; Zhou, N.; Li, L.; Huang, W.; Wang, D.; Zhang, C. In vivo and in vitro studies of borate based glass micro-fibers for dermal repairing. Mater. Sci. Eng. C 2016, 60, 437–445. [Google Scholar] [CrossRef]
- Llah, N.A.; Jamaludin, S.B.; Daud, Z.C.; Zaludin, M.A.F.; Jamal, Z.A.Z.; Idris, M.S.; Osman, R.A.M. Corrosion behavior of Mg-3Zn/bioglass (45S5) composite in simulated body fluid (SBF) and phosphate buffered saline (PBS) solution. AIP Conf. Proc. 2016, 1756, 030001. [Google Scholar] [CrossRef]
- Kim, H.M.; Kishimoto, K.; Miyaji, F.; Kokubo, T.; Yao, T.; Suetsugu, Y.; Tanaka, J.; Nakamura, T. Composition and structure of apatite formed on organic polymer in simulated body fluid with a high content of carbonate ion. J. Mater. Sci. Mater. Med. 2000, 11, 421–426. [Google Scholar] [CrossRef] [PubMed]
- Oyane, A.; Kim, H.M.; Furuya, T.; Kokubo, T.; Miyazaki, T.; Nakamura, T. Preparation and assessment of revised simulated body fluids. J. Biomed. Mater. Res. A 2003, 65, 188–195. [Google Scholar] [CrossRef]
- Müller, L.; Müller, F.A. Preparation of SBF with different HCO3− content and its influence on the composition of biomimetic apatites. Acta Biomater. 2006, 2, 181–189. [Google Scholar] [CrossRef] [PubMed]
- Mei, J.; Shelton, R.M.; Marquis, P.M. Changes in the elemental composition of Bioglass during its surface development in the presence or absence of proteins. J. Mater. Sci. Mater. Med. 1995, 6, 703–707. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Rohanová, D.; Horkavcová, D.; Paidere, L.; Boccaccini, A.R.; Bozděchová, P.; Bezdička, P. Interaction of HEPES buffer with glass-ceramic scaffold: Can HEPES replace TRIS in SBF? J. Biomed. Mater. Res. B Appl. Biomater. 2018, 106, 143–152. [Google Scholar] [CrossRef]
- Horkavcová, D.; Rohanová, D.; Stříbny, A.; Schuhladen, K.; Boccaccini, A.R.; Bezdička, P. Interaction of MOPS buffer with glass–ceramic scaffold: Effect of (PO4)3− ions in SBF on kinetics and morphology of formatted hydroxyapatite. J. Biomed. Mater. Res. B Appl. Biomater. 2020, 108, 1888–1896. [Google Scholar] [CrossRef]
- Bohner, M.; Lemaitre, J. Can bioactivity be tested in vitro with SBF solution? Biomaterials 2009, 30, 2175–2179. [Google Scholar] [CrossRef]
- Baino, F.; Yamaguchi, S. The use of simulated body fluid (SBF) for assessing materials bioactivity in the context of tissue engineering: Review and challenge. Biomimetics 2020, 5, 57. [Google Scholar] [CrossRef]
- Zhang, D.; Hupa, M.; Aro, H.T.; Hupa, L. Influence of fluid circulation on in vitro reactivity of bioactive glass particles. Mater. Chem. Phys. 2008, 111, 497–502. [Google Scholar] [CrossRef]
- Rámila, A.; Vallet-Regí, M. Static and dynamic in vitro study of a sol–gel glass bioactivity. Biomaterials 2001, 22, 2301–2306. [Google Scholar] [CrossRef] [PubMed]
- Kang, Y.; Xu, X.; Yin, G.; Chen, A.; Liao, L.; Yao, Y.; Huang, Z.; Liao, X. A comparative study of the in vitro degradation of poly(l-lactic acid)/β-tricalcium phosphate scaffold in static and dynamic simulated body fluid. Eur. Polym. J. 2007, 43, 1768–1778. [Google Scholar] [CrossRef]
- ISO 23317:2014; Implants for Surgery—In Vitro Evaluation for Apatite-Forming Ability of Implant Materials. Available online: https://www.iso.org/standard/65054.html?utm_source=chatgpt.com (accessed on 8 September 2025).
- Bellucci, D.; Salvatori, R.; Anesi, A.; Chiarini, L.; Cannillo, V. SBF assays, direct and indirect cell culture tests to evaluate the biological performance of bioglasses and bioglass-based composites: Three paradigmatic cases. Mater. Sci. Eng. C 2019, 96, 757–764. [Google Scholar] [CrossRef]
- Zadpoor, A.A. Relationship between in vitro apatite-forming ability measured using simulated body fluid and in vivo bioactivity of biomaterials. Mater. Sci. Eng. C 2014, 35, 134–143. [Google Scholar] [CrossRef] [PubMed]
- Ciraldo, F.E.; Boccardi, E.; Melli, V.; Westhauser, F.; Boccaccini, A.R. Tackling bioactive glass excessive in vitro bioreactivity: Preconditioning approaches for cell culture tests. Acta Biomater. 2018, 75, 3–10. [Google Scholar] [CrossRef]
- Varila, L.; Fagerlund, S.; Lehtonen, T.; Tuominen, J.; Hupa, L. Surface reactions of bioactive glasses in buffered solutions. J. Eur. Ceram. Soc. 2012, 32, 2757–2763. [Google Scholar] [CrossRef]
- Moorthi, A.; Saravanan, S.; Srinivasan, N.; Partridge, N.C.; Zhu, J.; Qin, L.; Selvamurugan, N. Synthesis, characterization and biological action of nano-bioglass ceramic particles for bone formation. J. Biomater. Tissue Eng. 2012, 2, 197–205. [Google Scholar] [CrossRef]
- Nayak, J.P.; Bera, J. Effect of sintering temperature on mechanical behaviour and bioactivity of sol-gel synthesized bioglass-ceramics using rice husk ash as a silica source. Appl. Surf. Sci. 2010, 257, 458–462. [Google Scholar] [CrossRef]
- Clupper, D.C.; Mecholsky, J.J.; Latorre, G.P.; Greenspan, D.C. Sintering temperature effects on the in vitro bioactive response of tape cast and sintered bioactive glass-ceramic in Tris buffer. J. Biomed. Mater. Res. 2001, 57, 532–540. [Google Scholar] [CrossRef] [PubMed]
- Kirste, G.; Brandt-Slowik, J.; Bocker, C.; Steinert, M.; Geiss, R.; Brauer, D.S. Effect of chloride ions in Tris buffer solution on bioactive glass apatite mineralization. Int. J. Appl. Glass. Sci. 2017, 8, 438–449. [Google Scholar] [CrossRef]
- Nommeots-Nomm, A.; Hupa, L.; Rohanová, D.; Brauer, D.S. A review of acellular immersion tests on bioactive glasses––influence of medium on ion release and apatite formation. Int. J. Appl. Glass Sci. 2020, 11, 537–551. [Google Scholar] [CrossRef]
- Wetzel, R.; Brauer, D.S. Apatite formation of substituted Bioglass 45S5: SBF vs. Tris. Mater. Lett 2019, 257, 126760. [Google Scholar] [CrossRef]
- Ogino, M.; Hench, L.L. Formation of calcium phosphate films on silicate glasses. J. Non-Cryst. Solids 1980, 38, 673–678. [Google Scholar] [CrossRef]
- Cerruti, M.; Bianchi, C.L.; Bonino, F.; Damin, A.; Perardi, A.; Morterra, C. Surface modifications of bioglass immersed in TRIS-buffered solution. A multitechnical spectroscopic study. J. Phys. Chem. B 2005, 109, 14496–14505. [Google Scholar] [CrossRef]
- Bellucci, D.; Bolelli, G.; Cannillo, V.; Cattini, A.; Sola, A. In situ Raman spectroscopy investigation of bioactive glass reactivity: Simulated body fluid solution vs TRIS-buffered solution. Mater. Charact. 2011, 62, 1021–1028. [Google Scholar] [CrossRef]
- Rohanová, D.; Boccaccini, A.R.; Yunos, D.M.; Horkavcová, D.; Březovská, I.; Helebrant, A. TRIS buffer in simulated body fluid distorts the assessment of glass-ceramic scaffold bioactivity. Acta Biomater. 2011, 7, 2623–2630. [Google Scholar] [CrossRef]
- Hlvác, J.; Rohanová, D.; Helebrant, A. The effect of Tris-buffer on the leaching behaviour of bioactive glass-ceramics. Ceram. Silik. 1994, 38, 119. [Google Scholar]
- Radin, S.; Ducheyne, P.; Rothman, B.; Conti, A. The effect of in vitro modeling conditions on the surface reactions of bioactive glass. J. Biomed. Mater. Res. 1997, 37, 363–375. Available online: https://pubmed.ncbi.nlm.nih.gov/9368141/ (accessed on 4 February 2025). [CrossRef]
- Fagerlund, S.; Hupa, L.; Hupa, M. Dissolution patterns of biocompatible glasses in 2-Amino-2-hydroxymethyl-propane-1,3-diol (Tris) buffer. Acta Biomater. 2013, 9, 5400–5410. [Google Scholar] [CrossRef] [PubMed]
- Hong, Z.; Reis, R.L.; Mano, J.F. Preparation and in vitro characterization of novel bioactive glass ceramic nanoparticles. J. Biomed. Mater. Res. A 2009, 88, 304–313. [Google Scholar] [CrossRef] [PubMed]
- Loh, Z.W.; Zaid, M.H.M.; Kechik, M.M.A.; Fen, Y.W.; Amin, K.M.; Cheong, W.M. New formulation calcium-based 45S5 bioactive glass: In vitro assessment in PBS solution for potential dental applications. J. Mater. Res. Technol. 2023, 24, 3815–3825. [Google Scholar] [CrossRef]
- Vienna, J.D.; Neeway, J.J.; Ryan, J.V.; Kerisit, S.N. Impacts of glass composition, pH, and temperature on glass forward dissolution rate. NPJ Mater. Degrad. 2018, 2, 22. [Google Scholar] [CrossRef]
- Zhou, Z.; Yi, Q.; Liu, X.; Liu, L.; Liu, Q. In vitro degradation behaviors of Poly-l-lactide/bioactive glass composite materials in phosphate-buffered solution. Polym. Bull. 2009, 63, 575–586. [Google Scholar] [CrossRef]
- Lusvardi, G.; Fraulini, F.; Cavazzoli, C.; Zambon, A. Evaluation of the behaviour of hydrogels containing mesoporous glasses doped with cerium and loaded with polyphenols. Ceram. Int. 2024, 50, 33937–33945. [Google Scholar] [CrossRef]
- Mehrabi, T.; Mesgar, A.S. In vitro biomineralization potential in simulated wound fluid and antibacterial efficacy of biologically-active glass nanoparticles containing B2O3/ZnO. Colloids Surf B Biointerfaces 2022, 212, 112338. [Google Scholar] [CrossRef]
- Overstreet, D.J.; Dutta, D.; Stabenfeldt, S.E.; Vernon, B.L. Injectable hydrogels. J. Polym. Sci. B Polym. Phys. 2012, 50, 881–903. [Google Scholar] [CrossRef]
- Fagerlund, S.; Hupa, L.; Hupa, M. Comparison of reactions of bioactive glasses in different aqueous solutions. Ceram. Trans. 2010, 218, 101–113. [Google Scholar] [CrossRef]
- Islam, M.T.; Felfel, R.M.; Neel, E.A.A.; Grant, D.M.; Ahmed, I.; Hossain, K.M.Z. Bioactive calcium phosphate–based glasses and ceramics and their biomedical applications: A review. J. Tissue Eng. 2017, 8, 2041731417719170. [Google Scholar] [CrossRef]
- Wu, C.; Xiao, Y. Evaluation of the In Vitro Bioactivity of Bioceramics. Bone Tissue Regen. Insights 2009, 2, 25–29. [Google Scholar] [CrossRef]
- Salètes, M.; Vartin, M.; Mocquot, C.; Chevalier, C.; Grosgogeat, B.; Colon, P.; Attik, N. Mesoporous bioactive glasses cytocompatibility assessment: A review of in vitro studies. Biomimetics 2021, 6, 9. [Google Scholar] [CrossRef]
- Goud, N.S. Biocompatibility Evaluation of Medical Devices. In A Comprehensive Guide to Toxicology in Nonclinical Drug Development; Faqi, A.S., Ed.; Elsevier: Amsterdam, The Netherlands, 2016; pp. 825–840. [Google Scholar] [CrossRef]
- Singh, S.; Singh, G.; Bala, N. Analysis of in vitro corrosion behavior and hemocompatibility of electrophoretically deposited bioglass–chitosan–iron oxide coating for biomedical applications. J. Mater. Res. 2020, 35, 1749–1761. [Google Scholar] [CrossRef]
- Sergi, R.; Bellucci, D.; Salvatori, R.; Cannillo, V. Chitosan-based bioactive glass gauze: Microstructural properties, in vitro bioactivity, and biological tests. Materials 2020, 13, 2819. [Google Scholar] [CrossRef]
- ISO 10993-12:2007; Biological Evaluation of Medical Devices—Part 12: Sample Preparation and Reference Materials. ISO: Geneva, Switzerland, 2007. Available online: https://www.iso.org/standard/40384.html (accessed on 8 April 2025).
- ISO 10993-5:2009; Biological Evaluation of Medical Devices—Part 5: Tests for In Vitro Cytotoxicity. ISO: Geneva, Switzerland, 2009. Available online: https://www.iso.org/standard/36406.html (accessed on 8 April 2025).
- Chraniuk, M.; Panasiuk, M.; Hovhannisyan, L.; Żołędowska, S.; Nidzworski, D.; Ciołek, L.; Woźniak, A.; Jaegermann, Z.; Biernat, M.; Gromadzka, B. The Preliminary Assessment of New Biomaterials Necessitates a Comparison of Direct and Indirect Cytotoxicity Methodological Approaches. Polymers 2022, 14, 4522. [Google Scholar] [CrossRef]
- LI, W.; ZHOU, J.; XU, Y. Study of the in vitro cytotoxicity testing of medical devices. Biomed. Rep. 2015, 3, 617–620. [Google Scholar] [CrossRef]
- Bellucci, D.; Sola, A.; Salvatori, R.; Anesi, A.; Chiarini, L.; Cannillo, V. Sol-gel derived bioactive glasses with low tendency to crystallize: Synthesis, post-sintering bioactivity and possible application for the production of porous scaffolds. Mater. Sci. Eng. C 2014, 43, 573–586. [Google Scholar] [CrossRef] [PubMed]
- Fernandes, G.V.O.; Alves, G.G.; Linhares, A.B.R.; Da Silva, M.H.P.; Granjeiro, J.M. Evaluation of cytocompatibility of bioglass-niobium granules with human primary osteoblasts: A multiparametric approach. In Key Engineering Materials; Trans Tech Publications Ltd: Baech, Switzerland, 2012; pp. 37–42. [Google Scholar] [CrossRef]
- Strober, W. Trypan blue exclusion test of cell viability. Curr. Protoc. Immunol. 2001, 21, A-3B. [Google Scholar] [CrossRef]
- Deb, S.; Mandegaran, R.; Di Silvio, L. A porous scaffold for bone tissue engineering/45S5 Bioglass® derived porous scaffolds for co-culturing osteoblasts and endothelial cells. J. Mater. Sci. Mater. Med. 2010, 21, 893–905. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Ye, X.J.; Li, J.S. Preparation and biocompatibility evaluation of apatite/wollastonite-derived porous bioactive glass ceramic scaffolds. Biomed. Mater. 2009, 4, 045007. [Google Scholar] [CrossRef]
- Van De Loosdrecht, A.A.; Nennie, E.; Ossenkoppele, G.J.; Beelen, R.H.J.; Langenhuijsen, M.M.A.C. Cell mediated cytotoxicity against U 937 cells by human monocytes and macrophages in a modified colorimetric A methodological study. J. Immunol. Methods 1991, 141, 15–22. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.D.; Abudhahir, K.M.; Selvamurugan, N.; Vimalraj, S.; Murugesan, R.; Srinivasan, N.; Moorthi, A. Formulation and biological actions of nano-bioglass ceramic particles doped with Calcarea phosphorica for bone tissue engineering. Mater. Sci. Eng. C 2018, 83, 202–209. [Google Scholar] [CrossRef]
- Spirandeli, B.R.; Ribas, R.G.; Amaral, S.S.; Martins, E.F.; Esposito, E.; Vasconcellos, L.M.R.; Campos, T.M.B.; Thim, G.P.; Trichês, E.S. Incorporation of 45S5 bioglass via sol-gel in β-TCP scaffolds: Bioactivity and antimicrobial activity evaluation. Mater. Sci. Eng. C 2021, 131, 112453. [Google Scholar] [CrossRef]
- Hesaraki, S.; Barounian, M.H.; Farhangdoust, S.; Khorami, M.; Zamanian, A.; Borhan, S. Mechanical and In Vitro Biological Properties of Hydroxyapatite Bioceramics Reinforced with Strontium-Containing Nano-Bioactive Glass. Curr. Nanosci. 2012, 8, 612–622. [Google Scholar] [CrossRef]
- da Fonseca, G.F.; de Avelino, O.S.; de Mello, D.C.R.; do Prado, R.F.; Campos, T.M.B.; de Vasconcellos, L.M.R.; de Sousa Trichês, E.; Borges, A.L.S. Scaffolds of PCL combined to bioglass: Synthesis, characterization and biological performance. J. Mater. Sci. Mater. Med. 2020, 31, 41. [Google Scholar] [CrossRef]
- Kaczmarek, M.; Jurczyk, M.U.; Rubis, B.; Banaszak, A.; Kolecka, A.; Paszel, A.; Jurczyk, K.; Murias, M.; Sikora, J.; Jurczyk, M. In vitro biocompatibility of Ti-45S5 bioglass nanocomposites and their scaffolds. J. Biomed. Mater. Res. A 2014, 102, 1316–1324. [Google Scholar] [CrossRef] [PubMed]
- Morgan, D.M. Tetrazolium (MTT) Assay for Cellular Viability and Activity. Methods. Mol. Biol. 1998, 79, 179–184. [Google Scholar] [CrossRef]
- Sylvester, P.W. Optimization of the Tetrazolium Dye (MTT) Colorimetric Assay for Cellular Growth and Viability. Methods Mol. Biol. 2011, 716, 157–168. [Google Scholar] [CrossRef]
- Fooladi, A.A.I.; Hosseini, H.M.; Hafezi, F.; Hosseinnejad, F.; Nourani, M.R. Sol-gel-derived bioactive glass containing SiO2-MgO-CaO-P2O5 as an antibacterial scaffold. J. Biomed. Mater. Res. A 2013, 101, 1582–1587. [Google Scholar] [CrossRef] [PubMed]
- De-Deus, G.; Canabarro, A.; Alves, G.; Linhares, A.; Senne, M.I.; Granjeiro, J.M. Optimal Cytocompatibility of a Bioceramic Nanoparticulate Cement in Primary Human Mesenchymal Cells. J. Endod. 2009, 35, 1387–1390. [Google Scholar] [CrossRef]
- Tripathi, H.; Hira, S.K.; Kumar, A.S.; Gupta, U.; Manna, P.P.; Singh, S.P. Structural characterization and in vitro bioactivity assessment of SiO2–CaO–P2O5–K2O–Al2O3 glass as bioactive ceramic material. Ceram. Int. 2015, 41, 11756–11769. [Google Scholar] [CrossRef]
- Mitri, F.; Alves, G.; Fernandes, G.; König, B.; Rossi, A.; Granjeiro, J. Cytocompatibility of Porous Biphasic Calcium Phosphate Granules with Human Mesenchymal Cells by a Multiparametric Assay. Artif. Organs 2012, 36, 535–542. [Google Scholar] [CrossRef]
- Deliormanli, A.M. Preparation, in vitro mineralization and osteoblast cell response of electrospun 13–93 bioactive glass nanofibers. Mater. Sci. Eng. C 2015, 53, 262–271. [Google Scholar] [CrossRef]
- Funk, D.; Schrenk, H.H.; Frei, E. Serum albumin leads to false-positive results in the XTT and the MTT assay. Biotechniques 2007, 43, 178–186. [Google Scholar] [CrossRef] [PubMed]
- Roehm, N.W.; Rodgers, G.H.; Hatfield, S.M.; Glasebrook, A.L. An improved colorimetric assay for cell proliferation and viability utilizing the tetrazolium salt XTT. J. Immunol. Methods 1991, 142, 257–265. [Google Scholar] [CrossRef] [PubMed]
- Vasconcelos, E.V.; da Luz, F.B.; da Paz, S.P.A.; Reis, M.A.L.D.; da Silva, A.C.R.; Passos, M.F.; Barboza, C.A.G.; Monteiro, S.N.; Candido, V.S. Nanostructured 3D bioprinting of PLA with bioglass-CNT scaffolds for osseus tissue graft manufacturing. J. Mater. Res. Technol. 2023, 23, 5923–5938. [Google Scholar] [CrossRef]
- Chen, Q.Z.; Xu, J.L.; Yu, L.G.; Fang, X.Y.; Khor, K.A. Spark plasma sintering of sol-gel derived 45S5 Bioglass®-ceramics: Mechanical properties and biocompatibility evaluation. Mater. Sci. Eng. C 2012, 32, 494–502. [Google Scholar] [CrossRef]
- O’brien, J.; Wilson, I.; Orton, T.; Pognan, F.Ë.O. Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. Eur. J. Biochem. 2000, 267, 5421–5426. [Google Scholar] [CrossRef] [PubMed]
- Chen, Q.Z.; Efthymiou, A.; Salih, V.; Boccaccini, A.R. Bioglass®-derived glass-ceramic scaffolds: Study of cell proliferation and scaffold degradation in vitro. J. Biomed. Mater. Res. A 2008, 84, 1049–1060. [Google Scholar] [CrossRef]
- Longhin, E.M.; El Yamani, N.; Rundén-Pran, E.; Dusinska, M. The alamar blue assay in the context of safety testing of nanomaterials. Front. Toxicol. 2022, 4, 981701. [Google Scholar] [CrossRef]
- Ibrahim, S.; Marzuke, M.A.; Hussin, Z.H.; Bashah, N.S.K. In Vitro Cytocompatibility of Synthetic Calcium Phosphate Powder on L929 Fibroblast Cell. J. Ind. Technol. 2015, 23, 15–24. [Google Scholar] [CrossRef]
- Rampersad, S.N. Multiple Applications of Alamar Blue as an Indicator of Metabolic Function and Cellular Health in Cell Viability Bioassays. Sensors 2012, 12, 12347–12360. [Google Scholar] [CrossRef]
- Habibe, A.F.; Maeda, L.D.; Souza, R.C.; Barboza, M.J.R.; Daguano, J.K.M.F.; Rogero, S.O.; Santos, C. Effect of bioglass additions on the sintering of Y-TZP bioceramics. Mater. Sci. Eng. C 2009, 29, 1959–1964. [Google Scholar] [CrossRef]
- Ciapetti, G.; Granchi, D.; Stea, S.; Savarino, L.; Verri, E.; Gori, A.; Savioli, F.; Montanaro, L. Cytotoxicity testing of materials with limited in vivo exposure is affected by the duration of cell-material contact. J. Biomed. Mater. Res. 1998, 42, 485–490. [Google Scholar] [CrossRef]
- Altman, S.A.; Randers, L.; Rao, G. Comparison of Trypan Blue Dye Exclusion and Fluorometric Assays for Mammalian Cell Viability Determinations. Biotechnol. Prog. 1993, 9, 671–674. [Google Scholar] [CrossRef]
- Vitale-Brovarone, C.; Verné, E.; Robiglio, L.; Appendino, P.; Bassi, F.; Martinasso, G.; Muzio, G.; Canuto, R. Development of glass–ceramic scaffolds for bone tissue engineering: Characterisation, proliferation of human osteoblasts and nodule formation. Acta Biomater. 2007, 3, 199–208. [Google Scholar] [CrossRef]
- Bellucci, D.; Veronesi, E.; Dominici, M.; Cannillo, V. A new bioactive glass with extremely high crystallization temperature and outstanding biological performance. Mater. Sci. Eng. C 2020, 110, 110699. [Google Scholar] [CrossRef]
- Bachar, A.; Mabrouk, A.; Amrousse, R.; Azat, S.; Follet, C.; Mercier, C.; Bouchart, F. Properties, Bioactivity and Viability of the New Generation of Oxyfluoronitride Bioglasses. Eurasian Chem.-Technol. J. 2024, 26, 43–52. [Google Scholar] [CrossRef]
- Feoktistova, M.; Geserick, P.; Leverkus, M. Crystal violet assay for determining viability of cultured cells. Cold Spring Harb. Protoc. 2016, 2016, 343–346. [Google Scholar] [CrossRef] [PubMed]
- Tavares, D.D.S.; Castro, L.D.O.; de Almeida Soares, G.D.; Alves, G.G.; Granjeiro, J.M. Synthesis and cytotoxicity evaluation of granular magnesium substituted β-tricalcium phosphate. J. Appl. Oral Sci. 2013, 21, 37. [Google Scholar] [CrossRef]
- Pires, L.A.; de Meira, C.R.; Tokuhara, C.K.; de Oliveira, F.A.; Dainezi, V.B.; Graeff, M.S.Z.; Fortulan, C.A.; de Oliveira, R.C.; Puppin-Rontani, R.M.; Borges, A.F.S. Wettability and pre-osteoblastic behavior evaluations of a dense bovine hydroxyapatite ceramics. J. Oral Sci. 2020, 62, 259–264. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Gao, X.; Gong, W.; Zhang, Z.; Chen, X.; Dong, Y. Odontogenic differentiation and dentin formation of dental pulp cells under nanobioactive glass induction. Acta Biomater. 2014, 10, 2792–2803. [Google Scholar] [CrossRef]
- Mocquot, C.; Colon, P.; Fernando, D.; Jackson, P.; Pradelle-Plasse, N.; Grosgogeat, B.; Attik, N. The influence of experimental bioactive glasses on pulp cells behavior in vitro. Dent. Mater. 2020, 36, 1322–1331. [Google Scholar] [CrossRef]
- Lusvardi, G.; Stabellini, F.S.; Salvatori, R. P2O5-free cerium containing glasses: Bioactivity and cytocompatibility evaluation. Materials 2019, 12, 3267. [Google Scholar] [CrossRef]
- Bellucci, D.; Sola, A.; Anesi, A.; Salvatori, R.; Chiarini, L.; Cannillo, V. Bioactive glass/hydroxyapatite composites: Mechanical properties and biological evaluation. Mater. Sci. Eng. C 2015, 51, 196–205. [Google Scholar] [CrossRef]
- Malavasi, G.; Salvatori, R.; Zambon, A.; Lusvardi, G.; Rigamonti, L.; Chiarini, L.; Anesi, A. Cytocompatibility of potential bioactive cerium-doped glasses based on 45S5. Materials 2019, 12, 594. [Google Scholar] [CrossRef] [PubMed]
- Ampatzis, K.; Dermon, C.R. Sex differences in adult cell proliferation within the zebrafish (Danio rerio) cerebellum. Eur. J. Neurosci. 2007, 25, 1030–1040. [Google Scholar] [CrossRef]
- Duque, A.; Rakic, P. Identification of Proliferating and Migrating Cells by BrdU and Other Thymidine Analogs: Benefits and Limitations. In Immunocytochemistry and Related Techniques; Humana Press: New York, NY, USA, 2015; pp. 123–139. [Google Scholar] [CrossRef]
- Anesi, A.; Malavasi, G.; Chiarini, L.; Salvatori, R.; Lusvardi, G. Cell proliferation to evaluate preliminarily the presence of enduring self-regenerative antioxidant activity in cerium doped bioactive glasses. Materials 2020, 13, 2297. [Google Scholar] [CrossRef] [PubMed]
- Mead, T.J.; Lefebvre, V. Proliferation Assays (BrdU and EdU) on Skeletal Tissue Sections. Methods Mol. Biol. 2014, 1130, 233. [Google Scholar] [CrossRef]
- Bielby, R.C.; Christodoulou, I.S.; Pryce, R.S.; Radford, W.J.P.; Hench, L.L.; Polak, J.M. Time- and concentration-dependent effects of dissolution products of 58S sol-gel bioactive glass on proliferation and differentiation of murine and human osteoblasts. Tissue Eng. 2004, 10, 1018–1026. [Google Scholar] [CrossRef]
- Silver, I.A.; Deas, J.; Erecińska, M. Interactions of bioactive glasses with osteoblasts in vitro: Effects of 45S5 Bioglass®, and 58S and 77S bioactive glasses on metabolism, intracellular ion concentrations and cell viability. Biomaterials 2001, 22, 175–185. [Google Scholar] [CrossRef]
- Salic, A.; Mitchison, T.J. A chemical method for fast and sensitive detection of DNA synthesis in vivo. Proc. Natl. Acad. Sci. USA 2008, 105, 2415–2420. [Google Scholar] [CrossRef]
- Zeng, C.; Pan, F.; Jones, L.A.; Lim, M.M.; Griffin, E.A.; Sheline, Y.I.; Mintun, M.A.; Holtzman, D.M.; Mach, R.H. Evaluation of 5-ethynyl-2′-deoxyuridine staining as a sensitive and reliable method for studying cell proliferation in the adult nervous system. Brain Res. 2010, 1319, 21–32. [Google Scholar] [CrossRef]
- Kotogány, E.; Dudits, D.; Horváth, G.V.; Ayaydin, F. A rapid and robust assay for detection of S-phase cell cycle progression in plant cells and tissues by using ethynyl deoxyuridine. Plant Methods 2010, 6, 5. [Google Scholar] [CrossRef] [PubMed]
- Buck, S.B.; Bradford, J.; Gee, K.R.; Agnew, B.J.; Clarke, S.T.; Salic, A. Detection of S-phase Cell Cycle Progression using 5-ethynyl-2′-deoxyuridine Incorporation with Click Chemistry, an Alternative to using 5-bromo-2′-deoxyuridine Antibodies. Biotechniques 2008, 44, 927–929. [Google Scholar] [CrossRef] [PubMed]
- Wheeler, T.S.; Sbravati, N.D.; Janorkar, A.V. Mechanical & cell culture properties of elastin-like polypeptide, collagen, bioglass, and carbon nanosphere composites. Ann. Biomed. Eng. 2013, 41, 2042–2055. [Google Scholar] [CrossRef]
- Mosaddada, S.A.; Yazdaniana, M.; Tebyanian, H.; Tahmasebia, E.; Yazdanianb, A.; Seifalianc, A.; Tavakolizadehd, M. Fabrication and properties of developed collagen/strontium-doped Bioglass scaffolds for bone tissue engineering. J. Mater. Res. Technol. 2020, 9, 14799–14817. [Google Scholar] [CrossRef]
- Zhao, H.; Liang, G.; Liang, W.; Li, Q.; Huang, B.; Li, A.; Qiu, D.; Jin, D. In vitro and in vivo evaluation of the pH-neutral bioactive glass as high performance bone grafts. Mater. Sci. Eng. C 2020, 116, 111249. [Google Scholar] [CrossRef]
- Bernar, A.; Gebetsberger, J.V.; Bauer, M.; Streif, W.; Schirmer, M. Optimization of the Alizarin Red S Assay by Enhancing Mineralization of Osteoblasts. Int. J. Mol. Sci. 2023, 24, 723. [Google Scholar] [CrossRef] [PubMed]
- Vargas, G.E.; Mesones, R.V.; Bretcanu, O.; López, J.M.P.; Boccaccini, A.R.; Gorustovich, A. Biocompatibility and bone mineralization potential of 45S5 Bioglass®-derived glass-ceramic scaffolds in chick embryos. Acta Biomater. 2009, 5, 374–380. [Google Scholar] [CrossRef]
- Thomas, A.; Bera, J. Preparation and characterization of gelatin-bioactive glass ceramic scaffolds for bone tissue engineering. J. Biomater. Sci. Polym. Ed. 2019, 30, 561–579. [Google Scholar] [CrossRef]
- Kleinfehn, A.P.; Lindemann, J.A.L.; Razvi, A.; Philip, P.; Richardson, K.; Nettleton, K.; Becker, M.L.; Dean, D. Modulating Bioglass Concentration in 3D Printed Poly(propylene fumarate) Scaffolds for Post-Printing Functionalization with Bioactive Functional Groups. Biomacromolecules 2019, 20, 4345–4352. [Google Scholar] [CrossRef]
- Tahmasebifar, A.; Baran, E.T.; Yilmaz, B.; Pazarceviren, A.E. Bioprinting of bioglass-alginate/carboxymethyl cellulose for bone tissue engineering. Bioprinting 2023, 34, e00296. [Google Scholar] [CrossRef]
- Shankhwar, N.; Kumar, M.; Mandal, B.B.; Srinivasan, A. Novel polyvinyl alcohol-bioglass 45S5 based composite nanofibrous membranes as bone scaffolds. Mater. Sci. Eng. C 2016, 69, 1167–1174. [Google Scholar] [CrossRef]
- El-Sayed, M.M.H.; Mostafa, A.A.; Gaafar, A.M.; El Hotaby, W.; Hamzawy, E.M.A.; El-Okaily, M.S.; Gamal-Eldeen, A.M. In vitro kinetic investigations on the bioactivity and cytocompatibility of bioactive glasses prepared via melting and sol–gel techniques for bone regeneration applications. Biomed. Mater. 2017, 12, 015029. [Google Scholar] [CrossRef]
- Tang, Z.; Chen, H.; He, H.; Ma, C. Assays for alkaline phosphatase activity: Progress and prospects. TrAC Trends Anal. Chem. 2019, 113, 32–43. [Google Scholar] [CrossRef]
- Zhu, N.; Chatzistavrou, X.; Ge, L.; Qin, M.; Papagerakis, P.; Wang, Y. Biological properties of modified bioactive glass on dental pulp cells. J. Dent. 2019, 83, 18–26. [Google Scholar] [CrossRef]
- Morales-Hernandez, D.G.; Genetos, D.C.; Working, D.M.; Murphy, K.C.; Leach, J.K. Ceramic Identity Contributes to Mechanical Properties and Osteoblast Behavior on Macroporous Composite Scaffolds. J. Funct. Biomater. 2012, 3, 382–397. [Google Scholar] [CrossRef] [PubMed]
- Chi, C.Y.; Chen, C.Y.; Huang, J.Y.; Kuan, C.Y.; Lin, Y.Y.; Li, C.H.; Yang, C.C.; Lin, F.H. Preparation and in-vitro evaluation of Fe2O3-doped DP-bioglass in combination with 3D-printing and selective laser sintering process (3DP-SLS) for alveolar bone augmentation. Ceram. Int. 2021, 47, 12725–12734. [Google Scholar] [CrossRef]
- Saino, E.; Grandi, S.; Quartarone, E.; Maliardi, V.; Galli, D.; Bloise, N.; Fassina, L.; de Angelis, M.G.C.; Mustarelli, P.; Imbriani, M.; et al. In vitro calcified matrix deposition by human osteoblasts onto a zinc-containing bioactive glass. Eur. Cell. Mater. 2011, 21, 59–72. [Google Scholar] [CrossRef] [PubMed]
- Khoroushi, M.; Foroughi, M.R.; Karbasi, S.; Hashemibeni, B.; Khademi, A.A. Effect of Polyhydroxybutyrate/Chitosan/Bioglass nanofiber scaffold on proliferation and differentiation of stem cells from human exfoliated deciduous teeth into odontoblast-like cells. Mater. Sci. Eng. C 2018, 89, 128–139. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Yuan, Y.; Fang, L.; Xuan, Y. Promotion of osteogenesis by bioactive glass–ceramic coating: Possible involvement of the Hedgehog signaling pathway. J. Orthop. Sci. 2019, 24, 731–736. [Google Scholar] [CrossRef]
- Papynov, E.K.; Shichalin, O.O.; Belov, A.A.; Buravlev, I.Y.; Mayorov, V.Y.; Fedorets, A.N.; Buravleva, A.A.; Lembikov, A.O.; Gritsuk, D.V.; Kapustina, O.V.; et al. CaSiO3-HAp Metal-Reinforced Biocomposite Ceramics for Bone Tissue Engineering. J. Funct. Biomater. 2023, 14, 259. [Google Scholar] [CrossRef]
- Papynov, E.K.; Shichalin, O.O.; Kapustina, O.V.; Buravlev, I.Y.; Apanasevich, V.I.; Mayorov, V.Y.; Fedorets, A.N.; Lembikov, A.O.; Gritsuk, D.N.; Ovodova, A.V.; et al. Synthetic Calcium Silicate Biocomposite Based on Sea Urchin Skeleton for 5-Fluorouracil Cancer Delivery. Materials 2023, 16, 3495. [Google Scholar] [CrossRef] [PubMed]
- Ma, H.; Bell, K.N.; Loker, R.N. qPCR and qRT-PCR analysis: Regulatory points to consider when conducting biodistribution and vector shedding studies. Mol. Ther. Methods Clin. Dev. 2020, 20, 152. [Google Scholar] [CrossRef]
- Trandaș, A.N.; Trifan, A.; Ungureanu, A.D.; Dusciuc, A.; Opriș, M.; Milea, O.C.; Banciu, A.; Constantinoiu, I.; Busuioc, C.; Isopencu, G.O. Properties of europium and silver doped bioglass thin films obtained by two deposition methods: Biointerfaces for bioinert implants. Mater. Chem. Phys. 2023, 309, 128396. [Google Scholar] [CrossRef]
- Lavric, R.; Vreme, C.; Busuioc, C.; Isopencu, G.O.; Nicoara, A.I.; Oprea, O.C.; Banciu, D.D.; Constantinoiu, I.; Musat, A.M.R. The Effect of Silver and Samarium on the Properties of Bioglass Coatings Produced by Pulsed Laser Deposition and Spin Coating. J. Funct. Biomater. 2023, 14, 560. [Google Scholar] [CrossRef]
- Saino, E.; Maliardi, V.; Quartarone, E.; Fassina, L.; Benedetti, L.; Gabriella, M.; De Angelis, C.; Mustarelli, P.; Facchini, A.; Visai, L. In Vitro Enhancement of SAOS-2 Cell Calcified Matrix Deposition onto Radio Frequency Magnetron Sputtered Bioglass-Coated Titanium Scaffolds. Tissue Eng. Part A 2010, 16, 995–1008. [Google Scholar] [CrossRef]
- Zhang, K.; Chai, B.; Ji, H.; Chen, L.; Ma, Y.; Zhu, L.; Xu, J.; Wu, Y.; Lan, Y.; Li, H.; et al. Bioglass promotes wound healing by inhibiting endothelial cell pyroptosis through regulation of the connexin 43/reactive oxygen species (ROS) signaling pathway. Lab. Investig. 2022, 102, 90–101. [Google Scholar] [CrossRef]
- Li, Y.; Zhang, X.; He, D.; Ma, Z.; Xue, K.; Li, H. 45S5 Bioglass® works synergistically with siRNA to downregulate the expression of matrix metalloproteinase-9 in diabetic wounds. Acta Biomater. 2022, 145, 372–389. [Google Scholar] [CrossRef]
- Usseglio, J.; Dumur, A.; Pagès, E.; Renaudie, É.; Abélanet, A.; Brie, J.; Champion, É.; Magnaudeix, A. Microporous Hydroxyapatite-Based Ceramics Alter the Physiology of Endothelial Cells through Physical and Chemical Cues. J. Funct. Biomater. 2023, 14, 460. [Google Scholar] [CrossRef] [PubMed]
- Tavakoli, M.; Bateni, E.; Rismanchian, M.; Fathi, M.; Doostmohammadi, A.; Rabiei, A.; Sadeghi, H.; Etebari, M.; Mirian, M. Genotoxicity effects of nano bioactive glass and Novabone bioglass on gingival fibroblasts using single cell gel electrophoresis (comet assay): An in vitro study. Dent. Res. J. 2012, 9, 314–320. [Google Scholar]
- Kido, H.W.; Oliveira, P.; Parizotto, N.A.; Crovace, M.C.; Zanotto, E.D.; Peitl-Filho, O.; Fernandes, K.P.S.; Mesquita-Ferrari, R.A.; Ribeiro, D.A.; Renno, A.C.M. Histopathological, cytotoxicity and genotoxicity evaluation of Biosilicate® glass-ceramic scaffolds. J. Biomed. Mater. Res. A 2013, 101, 667–673. [Google Scholar] [CrossRef] [PubMed]
- Collins, A.R.; Oscoz, A.A.; Brunborg, G.; Gaivão, I.; Giovannelli, L.; Kruszewski, M.; Smith, C.C.; Štětina, R. The comet assay: Topical issues. Mutagenesis 2008, 23, 143–151. [Google Scholar] [CrossRef] [PubMed]
Characteristic | Static Methods | Dynamic Methods |
---|---|---|
Test conditions | Conditions are fixed and do not change during the experiment. | Conditions can vary during the experiment.
|
Advantages |
|
|
Disadvantages |
|
|
Ion Concentration (mM) | Human Blood Plasma | The Original SBF |
---|---|---|
Na+ | 142.0 | 142.0 |
K+ | 5.0 | 5.0 |
Mg2+ | 1.5 | 1.5 |
Ca2+ | 2.5 | 2.5 |
Cl− | 103.0 | 148.8 |
HCO3− | 27.0 | 4.2 |
HPO42− | 1.0 | 1.0 |
SO42− | 0.5 | 0 |
Ion Concentration (mM) | c-SBF | r-SBF | i-SBF | m-SBF |
---|---|---|---|---|
Na+ | 142.0 | 142.0 | 142.0 | 142.0 |
K+ | 5.0 | 5.0 | 5.0 | 5.0 |
Mg2+ | 1.5 | 1.5 | 1.5 | 1.5 |
Ca2+ | 2.5 | 2.5 | 1.6 | 2.5 |
Cl− | 147.8 | 103.0 | 103.0 | 103.0 |
HCO3− | 4.2 | 27.0 | 27.0 | 4.2 |
HPO42− | 1.0 | 1.0 | 1.0 | 1.0 |
SO42− | 0.5 | 0.5 | 0.5 | 0.5 |
Recommended Ranges | ||
---|---|---|
Mass-to-SBF volume ratio | BGs in fine powder or particulate form | |
1.5 mg/mL | Maçon et al. [57] | |
2 mg/mL | Pirayesh et al. [59] | |
1 mg/mL | Zheng et al. [62]; Bano et al. [63] | |
Surface area-to-SBF volume ratio | Bulk/disk samples follow ISO 23317:2014 [78] | |
Surface area/Volume of SBF = 0.4 cm−1 | Zhang et al. [75]; Mei et al. [69] | |
Surface area/Volume of SBF = 0.05 cm−1 | Müller et al. [68] |
Ion Concentration (mM) | Human Blood Plasma | TRIS |
---|---|---|
Na+ | 142.0 | - |
K+ | 5.0 | - |
Mg2+ | 1.5 | - |
Ca2+ | 2.5 | - |
Cl− | 103.0 | 45.0 |
HCO3− | 27.0 | - |
HPO42− | 1.0 | - |
SO42− | 0.5 | - |
Ion Concentration (mM) | Human Blood Plasma | PBS |
---|---|---|
Na+ | 142.0 | 157.0 |
K+ | 5.0 | - |
Mg2+ | 1.5 | - |
Ca2+ | 2.5 | - |
Cl− | 103.0 | 100.9 |
HCO3− | 27.0 | - |
HPO42− | 1.0 | 24.9 |
SO42− | 0.5 | - |
H2PO4− | - | 5.5 |
Solution | Main Composition | Buffering System | Advantages | Disadvantages |
---|---|---|---|---|
SBF | Inorganic ions, including Ca2+, PO43−, Cl−, Na+ | Tris-buffer | Promotes spontaneous HAp nucleation; ISO standardized for bulk samples; commonly accepted guidelines for testing BG powders | May overestimate bioactivity (false positives); sensitive to supersaturation and pH changes [46] |
TRIS | Tris(hydroxymethyl)aminomethane, Cl− | TRIS-HCl | Useful for studying ion release kinetics; no interference from external Ca/P | HAp formation is not supported if the material fails to release a sufficient amount of ions [103] |
PBS | NaCl, phosphate salts | Weak phosphate buffer | Chemically stable; suitable for long-term degradation studies [103] | Low reactivity; lacks Ca2+ |
SWF | NaCl, KCl. NaHCO3, NaH2PO4 | Bicarbonate-based | Designed for wound healing materials; mimics wound environment | Not standardized; limited comparative data; rarely used for ceramics |
Test on Extracts | Direct Methods | Indirect Methods | |
---|---|---|---|
Advantages |
|
|
|
Disadvantages |
|
|
|
Target Tissue | Cell Line Examples | Biological Parameter Assessed |
---|---|---|
Bone | MG-63, Saos-2, hMSC | Proliferation, differentiation, ALP activity |
Dentin | hMSCs | Mineralization, gene expression |
Soft tissue | Fibroblasts, endothelial cells | Adhesion, proliferation, cytotoxicity |
Assay | Purpose | Advantages | Disadvantages | False Positives/Negatives Considerations |
---|---|---|---|---|
Cytotoxicity and viability assays | ||||
MTT XTT Alamar Blue | Measure metabolic activity | Quantitative, user-friendly, compatible with various cell types | Potential interference from the turbidity of the material or solution |
|
NR | Lysosomal integrity (viability) | Simple, no extensive preparation needed | Sensitive to variations in cell density | False positive from dye uptake by dead/dying cells |
Trypan Blue | Cell membrane integrity | Simple, low cost | Requires manual counting; subjective | False positives if dying cells temporarily exclude the dye |
Crystal violet | Cell adhesion and density | Simple, low cost | Requires microscopy; fixation may alter morphology | False negatives for poor staining |
Proliferation assay | ||||
BrdU EdU | DNA synthesis and cell proliferation | High specificity, detects active cell division | Requires cell fixation and denaturation, which may alter morphology | False negatives if cells proliferate slowly or are in quiescence |
Differentiation assays | ||||
Alizarin Red | Visualize mineralization | Direct quantification of mineral deposits | Requires fixation; may alter morphology | False negatives for poor staining |
ALP activity assay | Osteogenic differentiation marker | Specific for osteoblastic activity; standard equipment | Time-point specific | False negatives if ALP expression has declined during later stages of differentiation |
Gene and protein expression assays | ||||
qPCR | Gene expression during differentiation | High sensitivity and specificity | Requires careful optimization | False negatives linked to dye fluorescence limitations |
Immunocytochemistry | Protein localization and cell morphology | High-resolution imaging of cell-material interactions | Labor-intensive; requires advanced microscopy | False positives from non-specific antibody binding |
Genotoxicity assay | ||||
Comet Assay | DNA damage detection | Sensitive to various DNA lesions | Complex; labor-intensive | False positives from transient DNA breaks |
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
De Micco, S.; Bellucci, D.; Cannillo, V. The Assessment of Bioactivity and Biological Responsiveness in Bioactive Glasses and Ceramics: A Review of Available Techniques. Materials 2025, 18, 4393. https://doi.org/10.3390/ma18184393
De Micco S, Bellucci D, Cannillo V. The Assessment of Bioactivity and Biological Responsiveness in Bioactive Glasses and Ceramics: A Review of Available Techniques. Materials. 2025; 18(18):4393. https://doi.org/10.3390/ma18184393
Chicago/Turabian StyleDe Micco, Simone, Devis Bellucci, and Valeria Cannillo. 2025. "The Assessment of Bioactivity and Biological Responsiveness in Bioactive Glasses and Ceramics: A Review of Available Techniques" Materials 18, no. 18: 4393. https://doi.org/10.3390/ma18184393
APA StyleDe Micco, S., Bellucci, D., & Cannillo, V. (2025). The Assessment of Bioactivity and Biological Responsiveness in Bioactive Glasses and Ceramics: A Review of Available Techniques. Materials, 18(18), 4393. https://doi.org/10.3390/ma18184393