Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,482)

Search Parameters:
Keywords = osteoblast activity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 3316 KiB  
Article
Evaluation of Collagenic Porcine Bone Blended with a Collagen Gel for Bone Regeneration: An In Vitro Study
by Tania Vanessa Pierfelice, Chiara Cinquini, Morena Petrini, Emira D’Amico, Camillo D’Arcangelo, Antonio Barone and Giovanna Iezzi
Int. J. Mol. Sci. 2025, 26(15), 7621; https://doi.org/10.3390/ijms26157621 - 6 Aug 2025
Abstract
A thermosensitive collagen-based gel (TSV gel), containing type I and III collagen, has been developed to improve the handling and stability of bone graft materials. However, its direct effect on osteoblasts is not well understood. This in vitro study evaluated the biological response [...] Read more.
A thermosensitive collagen-based gel (TSV gel), containing type I and III collagen, has been developed to improve the handling and stability of bone graft materials. However, its direct effect on osteoblasts is not well understood. This in vitro study evaluated the biological response of human oral osteoblasts to four bone substitutes: OsteoBiol® GTO® (larger granules with 20% TSV gel), Gen-OS® (smaller granules), Gen-OS® combined with 50% TSV gel (Gen-OS®+TSV), and TSV gel alone. Cell proliferation, adhesion, morphology, collagen and calcium deposition, alkaline phosphatase (ALP) activity, gene expression of osteogenic markers and integrins, and changes in pH and extracellular calcium and phosphate levels were investigated. All materials supported osteoblast activity, but Gen-OS+TSV and GTO showed the most pronounced effects. These two groups promoted better cell adhesion and proliferation, higher ALP activity, and greater matrix mineralization. GTO improved cell adhesion, while the addition of TSV gel to Gen-OS enhanced biological responses compared with Gen-OS alone. Integrins α2, α5, β1, and β3, important for cell attachment to collagen, were notably upregulated in Gen-OS+TSV and GTO. Both groups also showed increased expression of osteogenic markers such as BMP-2, ALP, and osteocalcin (OCN). Higher extracellular ion concentrations and a more alkaline pH were observed, particularly in conditions without cells, suggesting active ion uptake by osteoblasts. In conclusion, combining TSV gel with collagen-based granules improves the cellular environment for osteoblast activity and may support bone regeneration more effectively than using either component alone. Full article
(This article belongs to the Special Issue Molecular Studies of Bone Biology and Bone Tissue: 2nd Edition)
Show Figures

Graphical abstract

22 pages, 1078 KiB  
Review
The Cannabinoid Pharmacology of Bone Healing: Developments in Fusion Medicine
by Gabriel Urreola, Michael Le, Alan Harris, Jose A. Castillo, Augustine M. Saiz, Hania Shahzad, Allan R. Martin, Kee D. Kim, Safdar Khan and Richard Price
Biomedicines 2025, 13(8), 1891; https://doi.org/10.3390/biomedicines13081891 - 3 Aug 2025
Viewed by 404
Abstract
Background/Objectives: Cannabinoid use is rising among patients undergoing spinal fusion, yet its influence on bone healing is poorly defined. The endocannabinoid system (ECS)—through cannabinoid receptors 1 (CB1) and 2 (CB2)—modulates skeletal metabolism. We reviewed preclinical, mechanistic and clinical evidence to clarify how individual [...] Read more.
Background/Objectives: Cannabinoid use is rising among patients undergoing spinal fusion, yet its influence on bone healing is poorly defined. The endocannabinoid system (ECS)—through cannabinoid receptors 1 (CB1) and 2 (CB2)—modulates skeletal metabolism. We reviewed preclinical, mechanistic and clinical evidence to clarify how individual cannabinoids affect fracture repair and spinal arthrodesis. Methods: PubMed, Web of Science and Scopus were searched from inception to 31 May 2025 with the terms “cannabinoid”, “CB1”, “CB2”, “spinal fusion”, “fracture”, “osteoblast” and “osteoclast”. Animal studies, in vitro experiments and clinical reports that reported bone outcomes were eligible. Results: CB2 signaling was uniformly osteogenic. CB2-knockout mice developed high-turnover osteoporosis, whereas CB2 agonists (HU-308, JWH-133, HU-433, JWH-015) restored trabecular volume, enhanced osteoblast activity and strengthened fracture callus. Cannabidiol (CBD), a non-psychoactive phytocannabinoid with CB2 bias, accelerated early posterolateral fusion in rats and reduced the RANKL/OPG ratio without compromising final union. In contrast, sustained or high-dose Δ9-tetrahydrocannabinol (THC) activation of CB1 slowed chondrocyte hypertrophy, decreased mesenchymal-stromal-cell mineralization and correlated clinically with 6–10% lower bone-mineral density and a 1.8–3.6-fold higher pseudarthrosis or revision risk. Short-course or low-dose THC appeared skeletal neutral. Responses varied with sex, age and genetic background; no prospective trials defined safe perioperative dosing thresholds. Conclusions: CB2 activation and CBD consistently favor bone repair, whereas chronic high-THC exposure poses a modifiable risk for nonunion in spine surgery. Prospective, receptor-specific trials stratified by THC/CBD ratio, patient sex and ECS genotype are needed to establish evidence-based cannabinoid use in spinal fusion. Full article
(This article belongs to the Topic Cannabis, Cannabinoids and Its Derivatives)
Show Figures

Figure 1

16 pages, 948 KiB  
Review
Oxytocin: From Biomarker to Therapy for Postmenopausal Osteoporosis
by Tiago Franca, Joana Fonseca Ferreira, Melissa Mariana and Elisa Cairrao
Women 2025, 5(3), 27; https://doi.org/10.3390/women5030027 - 1 Aug 2025
Viewed by 142
Abstract
Postmenopausal osteoporosis is estrogen-dependent and results in an imbalance between bone formation and resorption. The approved therapy is intended to reduce the risk and consequences of fractures, but still has a number of contraindications and associated adverse effects. Recently, oxytocin has been shown [...] Read more.
Postmenopausal osteoporosis is estrogen-dependent and results in an imbalance between bone formation and resorption. The approved therapy is intended to reduce the risk and consequences of fractures, but still has a number of contraindications and associated adverse effects. Recently, oxytocin has been shown to have an anabolic effect on bone tissue, increasing the production of osteoblasts and inhibiting the activity of osteoclasts. Thus, this study aimed to examine the potential of oxytocin as a biomarker and therapeutic agent for postmenopausal osteoporosis. A PubMed search yielded 16 articles upon analysis of the inclusion and exclusion criteria. The results showed that, compared to women in the same age group without bone loss, those diagnosed with osteoporosis exhibited lower blood oxytocin levels, possibly related to a greater tendency towards fractures. The administration of oxytocin could be a promising strategy to enhance bone quality and, consequently, to reduce the incidence of fragility fractures; however, no human studies have been conducted regarding its use as a possible treatment. Thus, it is essential to increase the number of clinical trials in women with ovarian dysfunction and bone loss, in which oxytocin could become a viable therapeutic alternative. Full article
Show Figures

Figure 1

17 pages, 1015 KiB  
Review
Docosahexaenoic Acid Inhibits Osteoclastogenesis via FFAR4-Mediated Regulation of Inflammatory Cytokines
by Jinghan Ma, Hideki Kitaura, Fumitoshi Ohori, Aseel Marahleh, Ziqiu Fan, Angyi Lin, Kohei Narita, Kou Murakami and Hiroyasu Kanetaka
Molecules 2025, 30(15), 3180; https://doi.org/10.3390/molecules30153180 - 29 Jul 2025
Viewed by 301
Abstract
Osteoclastogenesis—the activation and differentiation of osteoclasts—is one of the pivotal processes of bone remodeling and is regulated by RANKL/RANK signaling, the decoy function of osteoprotegerin (OPG), and a cascade of pro- and anti-inflammatory cytokines. The disruption of this balance leads to pathological bone [...] Read more.
Osteoclastogenesis—the activation and differentiation of osteoclasts—is one of the pivotal processes of bone remodeling and is regulated by RANKL/RANK signaling, the decoy function of osteoprotegerin (OPG), and a cascade of pro- and anti-inflammatory cytokines. The disruption of this balance leads to pathological bone loss in diseases such as osteoporosis and rheumatoid arthritis. FFAR4 (Free Fatty Acid Receptor 4), a G protein-coupled receptor for long-chain omega-3 fatty acids, has been confirmed as a key mediator of metabolic and anti-inflammatory effects. This review focuses on how FFAR4 acts as the selective receptor for the omega-3 fatty acid docosahexaenoic acid (DHA). It activates two divergent signaling pathways. The Gαq-dependent cascade facilitates intracellular calcium mobilization and ERK1/2 activation. Meanwhile, β-arrestin-2 recruitment inhibits NF-κB. These collective actions reshape the cytokine environment. In macrophages, DHA–FFAR4 signaling lowers the levels of TNF-α, interleukin-6 (IL-6), and IL-1β while increasing IL-10 secretion. Consequently, the activation of NFATc1 and NF-κB p65 is profoundly suppressed under TNF-α or RANKL stimulation. Additionally, DHA modulates the RANKL/OPG axis in osteoblastic cells by suppressing RANKL expression, thereby reducing osteoclast differentiation in an inflammatory mouse model. Full article
Show Figures

Figure 1

9 pages, 635 KiB  
Article
Osteogenic Potential of Osteolforte: Gene and Protein-Level Evaluation in Human Bone Marrow Stromal Cells
by Da-Sol Kim, Soo-Kyung Bae, Yeon-Ju Kwak, Geum-Joung Youn and Hye-Ock Jang
Curr. Issues Mol. Biol. 2025, 47(8), 588; https://doi.org/10.3390/cimb47080588 - 24 Jul 2025
Viewed by 296
Abstract
Osteolforte, a compound with potential bone-regenerative properties, was investigated for its effects on human bone marrow stromal cells (hBMSCs). This study aimed to evaluate its impact on cell viability, osteogenic differentiation, and both gene and protein expression using a combination of assays, [...] Read more.
Osteolforte, a compound with potential bone-regenerative properties, was investigated for its effects on human bone marrow stromal cells (hBMSCs). This study aimed to evaluate its impact on cell viability, osteogenic differentiation, and both gene and protein expression using a combination of assays, including CCK-8, Alizarin Red S staining, Quantitative Real-Time PCR (qRT-PCR), and Western blot analysis. The results demonstrated that Osteolforte significantly enhanced osteogenic differentiation in hBMSCs. Alizarin Red S staining revealed increased mineralization, indicating elevated calcium deposition. Gene expression analysis showed an upregulation of key osteogenic markers, including runt-related transcription factor-2 (RUNX-2), collagen type I (COL-1), and bone morphogenetic protein-2 (BMP-2), supporting the role of Osteolforte in promoting osteoblastic activity. In particular, the elevated expression of RUNX-2—a master transcription factor in osteoblast differentiation along with COL-1, a major bone matrix component, and BMP-2, a key bone morphogenetic protein—highlights the compound’s osteogenic potential. In conclusion, Osteolforte enhances early-stage osteogenesis and mineralization in hBMSCs and represents a promising candidate for bone regeneration. Full article
Show Figures

Figure 1

16 pages, 2230 KiB  
Article
Three-Dimensional-Printed Biomimetic Scaffolds for Investigating Osteoblast-Like Cell Interactions in Simulated Microgravity: An In Vitro Platform for Bone Tissue Engineering Research
by Eleonora Zenobi, Giulia Gramigna, Elisa Scatena, Luca Panizza, Carlotta Achille, Raffaella Pecci, Annalisa Convertino, Costantino Del Gaudio, Antonella Lisi and Mario Ledda
J. Funct. Biomater. 2025, 16(8), 271; https://doi.org/10.3390/jfb16080271 - 24 Jul 2025
Viewed by 648
Abstract
Three-dimensional cell culture systems are relevant in vitro models for studying cellular behavior. In this regard, this present study investigates the interaction between human osteoblast-like cells and 3D-printed scaffolds mimicking physiological and osteoporotic bone structures under simulated microgravity conditions. The objective is to [...] Read more.
Three-dimensional cell culture systems are relevant in vitro models for studying cellular behavior. In this regard, this present study investigates the interaction between human osteoblast-like cells and 3D-printed scaffolds mimicking physiological and osteoporotic bone structures under simulated microgravity conditions. The objective is to assess the effects of scaffold architecture and dynamic culture conditions on cell adhesion, proliferation, and metabolic activity, with implications for osteoporosis research. Polylactic acid scaffolds with physiological (P) and osteoporotic-like (O) trabecular architectures were 3D-printed by means of fused deposition modeling technology. Morphometric characterization was performed using micro-computed tomography. Human osteoblast-like SAOS-2 and U2OS cells were cultured on the scaffolds under static and dynamic simulated microgravity conditions using a rotary cell culture system (RCCS). Scaffold biocompatibility, cell viability, adhesion, and metabolic activity were evaluated through Bromodeoxyuridine incorporation assays, a water-soluble tetrazolium salt assay, and an enzyme-linked immunosorbent assay of tumor necrosis factor-α secretion. Both scaffold models supported osteoblast-like cell adhesion and growth, with an approximately threefold increase in colonization observed on the high-porosity O scaffolds under dynamic conditions. The dynamic environment facilitated increased surface interaction, amplifying the effects of scaffold architecture on cell behavior. Overall, sustained cell growth and metabolic activity, together with the absence of detectable inflammatory responses, confirmed the biocompatibility of the system. Scaffold microstructure and dynamic culture conditions significantly influence osteoblast-like cell behavior. The combination of 3D-printed scaffolds and a RCCS bioreactor provides a promising platform for studying bone remodeling in osteoporosis and microgravity-induced bone loss. These findings may contribute to the development of advanced in vitro models for biomedical research and potential countermeasures for bone degeneration. Full article
(This article belongs to the Special Issue Functional Biomaterial for Bone Regeneration)
Show Figures

Graphical abstract

31 pages, 1902 KiB  
Review
Effects of Epigallocatechin-3-O-Gallate on Bone Health
by Patrycja Wróbel, Beata Czarczynska-Goslinska, Kyrylo Chornovolenko, Julia Liwarska, Jakub Kubiak, Tomasz Koczorowski, Agnieszka Malinska, Tomasz Goslinski and Magdalena Waszyk-Nowaczyk
Appl. Sci. 2025, 15(15), 8182; https://doi.org/10.3390/app15158182 - 23 Jul 2025
Viewed by 219
Abstract
Tea is one of the most consumed beverages in the world, belonging to the category of compounds known as tannins and flavonoids. One of the polyphenols found in large amounts in green tea leaves (Camellia sinensis) is epigallocatechin-3-O-gallate (EGCG). [...] Read more.
Tea is one of the most consumed beverages in the world, belonging to the category of compounds known as tannins and flavonoids. One of the polyphenols found in large amounts in green tea leaves (Camellia sinensis) is epigallocatechin-3-O-gallate (EGCG). Though EGCG has shown some pharmacological effects, to date, it has not been utilised as a therapeutic agent. This is attributed to the fact that EGCG lacks adequate stability, and it is known to degrade through epimerization or auto-oxidation processes, especially when it is exposed to light, temperature fluctuations, some pH values, or the presence of oxygen. Consuming green tea with EGCG can alleviate the effects of bone diseases, such as osteoporosis, and support faster bone regeneration in the case of fractures. Therefore, this review focuses on the current state of research, highlighting the effects of EGCG on bone biology, such as enhancing osteoblast differentiation, promoting bone mineralisation, improving bone microarchitecture, and inhibiting osteoclastogenesis through the modulation of the RANK/RANKL/OPG pathway. Additionally, EGCG exerts antioxidant, anti-inflammatory, and dose-dependent effects on bone cells. It also downregulates inflammatory markers (TNF-α, IL-1β, and COX-2) and reduces oxidative stress via the inhibition of reactive oxygen species generation and the activation of protective signalling pathways (e.g., MAPK and NF-κB). Studies in animal models confirm that EGCG supplementation leads to increased bone mass and strength. These findings collectively support the further exploration of EGCG as an adjunct in the treatment and prevention of metabolic bone diseases. The authors aim to present the relationship between EGCG and bone health, highlighting issues for future research and clinical applications. Full article
Show Figures

Figure 1

11 pages, 243 KiB  
Review
Adipokines: Do They Affect the Osteochondral Unit?
by Sergio Rosini, Gianantonio Saviola, Stefano Rosini, Eleonora Baldissarro and Luigi Molfetta
Rheumato 2025, 5(3), 9; https://doi.org/10.3390/rheumato5030009 - 22 Jul 2025
Viewed by 202
Abstract
Obesity, characterized by excessive or abnormal accumulation of body fat, is associated with a range of metabolic and inflammatory diseases, including osteoarthritis (OA). In obese individuals, adipose tissue expansion—via adipocyte hypertrophy or hyperplasia—is accompanied by altered secretion of adipokines such as leptin and [...] Read more.
Obesity, characterized by excessive or abnormal accumulation of body fat, is associated with a range of metabolic and inflammatory diseases, including osteoarthritis (OA). In obese individuals, adipose tissue expansion—via adipocyte hypertrophy or hyperplasia—is accompanied by altered secretion of adipokines such as leptin and adiponectin, which play significant roles in immune modulation, metabolism, and skeletal homeostasis. Leptin, acting through the hypothalamus, regulates the sympathetic nervous system and modulates hormonal axes, influencing bone metabolism and cartilage integrity. Elevated leptin concentrations in the synovial fluid, and the presence of its receptors on cartilage surfaces, suggest its direct role in cartilage degradation and OA progression. Conversely, adiponectin exerts anti-inflammatory effects, modulates osteoblast and macrophage activity, and appears to have a protective function in joint metabolism. These findings underscore the complex interplay between the adipose tissue, adipokines, and the osteochondral unit, highlighting the importance of their balance in maintaining joint health. Full article
29 pages, 15018 KiB  
Article
Investigating the Osteoregenerative Properties of Juglans regia L. Extract on Mesenchymal Stem Cells and Osteoblasts Through Evaluation of Bone Markers: A Pilot Study
by Alina Hanga-Fărcaș, Gabriela Adriana Filip, Simona Valeria Clichici, Laura Grațiela Vicaș, Olga Şoritău, Otilia Andercou, Luminița Fritea and Mariana Eugenia Mureșan
J. Funct. Biomater. 2025, 16(7), 268; https://doi.org/10.3390/jfb16070268 - 21 Jul 2025
Viewed by 485
Abstract
Bone tissue regeneration is a complex process that takes place at the level of osteoblasts derived from mesenchymal cells and occurs under the action of multiple signaling pathways and through the expression of osteoregenerative markers. The leaf extract of Juglans regia L. (JR) [...] Read more.
Bone tissue regeneration is a complex process that takes place at the level of osteoblasts derived from mesenchymal cells and occurs under the action of multiple signaling pathways and through the expression of osteoregenerative markers. The leaf extract of Juglans regia L. (JR) is rich in polyphenols with demonstrated osteoregeneration effects. In the present study, we investigated the extract’s effects on three types of cells with various stages of differentiation: adult mesenchymal stem cells (MSCs), osteoblasts at low passage (O6) and osteoblasts at advanced passage (O10). To assess the efficacy of the walnut leaf extract, in vitro treatments were performed in comparison with ellagic acid (EA) and catechin (CAT). The osteoregenerative properties of the leaf extract were evaluated in terms of cell viability, bone mineralization (by staining with alizarin red) and the expression of osteogenesis markers such as osteocalcin (OC), osteopontin (OPN), dentin matrix acidic phosphoprotein 1 (DMP1) and collagen type 1A. Another compound implicated in oxidative stress response, but also a bone homeostasis regulator, nuclear factor erythroid 2-related factor 2 (NRF2), was studied by immunocytochemistry. Together with collagen amount, alkaline phosphatase (ALP) activity and NF-kB levels were measured in cell lysates and supernatants. The obtained results demonstrate that JR treatment induced osteogenic differentiation and bone mineralization, and it showed protective effects against oxidative stress. Full article
(This article belongs to the Special Issue Functional Biomaterial for Bone Regeneration)
Show Figures

Figure 1

20 pages, 12298 KiB  
Article
Impact of Metastatic Microenvironment on Physiology and Metabolism of Small Cell Neuroendocrine Prostate Cancer Patient-Derived Xenografts
by Shubhangi Agarwal, Deepti Upadhyay, Jinny Sun, Emilie Decavel-Bueff, Robert A. Bok, Romelyn Delos Santos, Said Al Muzhahimi, Rosalie Nolley, Jason Crane, John Kurhanewicz, Donna M. Peehl and Renuka Sriram
Cancers 2025, 17(14), 2385; https://doi.org/10.3390/cancers17142385 - 18 Jul 2025
Viewed by 442
Abstract
Background: Potent androgen receptor pathway inhibitors induce small cell neuroendocrine prostate cancer (SCNC), a highly aggressive subtype of metastatic androgen deprivation-resistant prostate cancer (ARPC) with limited treatment options and poor survival rates. Patients with metastases in the liver have a poor prognosis relative [...] Read more.
Background: Potent androgen receptor pathway inhibitors induce small cell neuroendocrine prostate cancer (SCNC), a highly aggressive subtype of metastatic androgen deprivation-resistant prostate cancer (ARPC) with limited treatment options and poor survival rates. Patients with metastases in the liver have a poor prognosis relative to those with bone metastases alone. The mechanisms that underlie the different behavior of ARPC in bone vs. liver may involve factors intrinsic to the tumor cell, tumor microenvironment, and/or systemic factors, and identifying these factors is critical to improved diagnosis and treatment of SCNC. Metabolic reprogramming is a fundamental strategy of tumor cells to colonize and proliferate in microenvironments distinct from the primary site. Understanding the metabolic plasticity of cancer cells may reveal novel approaches to imaging and treating metastases more effectively. Methods: Using magnetic resonance (MR) imaging and spectroscopy, we interrogated the physiological and metabolic characteristics of SCNC patient-derived xenografts (PDXs) propagated in the bone and liver, and used correlative biochemical, immunohistochemical, and transcriptomic measures to understand the biological underpinnings of the observed imaging metrics. Results: We found that the influence of the microenvironment on physiologic measures using MRI was variable among PDXs. However, the MR measure of glycolytic capacity in the liver using hyperpolarized 13C pyruvic acid recapitulated the enzyme activity (lactate dehydrogenase), cofactor (nicotinamide adenine dinucleotide), and stable isotope measures of fractional enrichment of lactate. While in the bone, the congruence of the glycolytic components was lost and potentially weighted by the interaction of cancer cells with osteoclasts/osteoblasts. Conclusion: While there was little impact of microenvironmental factors on metabolism, the physiological measures (cellularity and perfusion) are highly variable and necessitate the use of combined hyperpolarized 13C MRI and multiparametric (anatomic, diffusion-, and perfusion- weighted) 1H MRI to better characterize pre-treatment tumor characteristics, which will be crucial to evaluate treatment response. Full article
(This article belongs to the Special Issue Magnetic Resonance in Cancer Research)
Show Figures

Figure 1

16 pages, 6631 KiB  
Article
Green Tea Extract Containing Epigallocatechin-3-Gallate Facilitates Bone Formation and Mineralization by Alleviating Iron-Overload-Induced Oxidative Stress in Human Osteoblast-like (MG-63) Cells
by Honghong Xu, Orawan Khantamat, Woranontee Korsieporn, Narisara Paradee, Jin Li, Yanping Zhong, Somdet Srichairatanakool and Pimpisid Koonyosying
Antioxidants 2025, 14(7), 874; https://doi.org/10.3390/antiox14070874 - 17 Jul 2025
Viewed by 355
Abstract
Secondary iron overload exacerbates osteoporosis by elevating reactive oxygen species (ROS), which suppress osteoblast function and enhance osteoclast activity, disrupting bone remodeling. Reducing iron overload and oxidative stress may improve bone health. Epigallocatechin-3-gallate (EGCG), the main bioactive compound in green tea extract (GTE), [...] Read more.
Secondary iron overload exacerbates osteoporosis by elevating reactive oxygen species (ROS), which suppress osteoblast function and enhance osteoclast activity, disrupting bone remodeling. Reducing iron overload and oxidative stress may improve bone health. Epigallocatechin-3-gallate (EGCG), the main bioactive compound in green tea extract (GTE), is recognized for its antioxidant and iron-chelating properties. This study examined the effect of GTE on bone formation and mineralization in iron-overloaded human osteoblast-like MG-63 cells. An iron-overloaded model was established using ferric ammonium citrate (FAC), followed by treatment with GTE, deferiprone (DFP), or their combination. GTE significantly reduced intracellular iron, ROS levels, and lipid peroxidation while upregulating the osteogenic marker BGLAP, the anti-resorptive marker OPG, and osteogenic mineralization, indicating restored bone health. These results suggest that EGCG-containing GTE mitigates iron-induced oxidative stress and promotes osteogenesis, highlighting its potential as a natural therapeutic supplement for managing iron-overload-associated osteoporosis. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Graphical abstract

29 pages, 2331 KiB  
Review
Therapeutic Potential of Tanshinones in Osteolytic Diseases: From Molecular and Cellular Pathways to Preclinical Models
by Rafael Scaf de Molon
Dent. J. 2025, 13(7), 309; https://doi.org/10.3390/dj13070309 - 9 Jul 2025
Viewed by 497
Abstract
Tanshinones are a class of lipophilic diterpenoid quinones extracted from Salvia miltiorrhiza (Dan shen), a widely used herb in traditional Chinese medicine. These compounds, particularly tanshinone IIA (T-IIA) and sodium tanshinone sulfonate (STS), have been acknowledged for their broad spectrum of biological activities, [...] Read more.
Tanshinones are a class of lipophilic diterpenoid quinones extracted from Salvia miltiorrhiza (Dan shen), a widely used herb in traditional Chinese medicine. These compounds, particularly tanshinone IIA (T-IIA) and sodium tanshinone sulfonate (STS), have been acknowledged for their broad spectrum of biological activities, including anti-inflammatory, antioxidant, anti-tumor, antiresorptive, and antimicrobial effects. Recent studies have highlighted the potential of tanshinones in the treatment of osteolytic diseases, characterized by excessive bone resorption, such as osteoporosis, rheumatoid arthritis, and periodontitis. The therapeutic effects of tanshinones in these diseases are primarily attributed to their ability to inhibit osteoclast differentiation and activity, suppress inflammatory cytokine production (e.g., tumor necrosis factor alpha (TNF-α), interleukin (IL)-1β, and IL-6), and modulate critical signaling pathways, including NF-kB, MAPK, PI3K/Akt, and the RANKL/RANK/OPG axis. Additionally, tanshinones promote osteoblast differentiation and mineralization by enhancing the expression of osteogenic markers such as Runx2, ALP, and OCN. Preclinical models have demonstrated that T-IIA and STS can significantly reduce bone destruction and inflammatory cell infiltration in arthritic joints and periodontal tissues while also enhancing bone microarchitecture in osteoporotic conditions. This review aims to provide a comprehensive overview of the pharmacological actions of tanshinones in osteolytic diseases, summarizing current experimental findings, elucidating underlying molecular mechanisms, and discussing the challenges and future directions for their clinical application as novel therapeutic agents in bone-related disorders, especially periodontitis. Despite promising in vitro and in vivo findings, clinical evidence remains limited, and further investigations are necessary to validate the efficacy, safety, and pharmacokinetics of tanshinones in human populations. Full article
(This article belongs to the Special Issue New Perspectives in Periodontology and Implant Dentistry)
Show Figures

Figure 1

20 pages, 3323 KiB  
Article
Determination of the Corrosion and Biocompatibility Properties of As-Cast TiNi Alloys
by Minja Miličić Lazić, Dijana Mitić, Biljana Dojčinović, Marko Lazić, Aleksandra S. Popović and Branimir N. Grgur
Metals 2025, 15(7), 758; https://doi.org/10.3390/met15070758 - 5 Jul 2025
Viewed by 239
Abstract
In this study, a TiNi alloy with a composition of 50 at.% of titanium and 50 at.% of nickel is investigated in terms of its corrosion and biocompatibility behavior for biomedical applications. The corrosion measurements, which include the determination of open-circuit potential and [...] Read more.
In this study, a TiNi alloy with a composition of 50 at.% of titanium and 50 at.% of nickel is investigated in terms of its corrosion and biocompatibility behavior for biomedical applications. The corrosion measurements, which include the determination of open-circuit potential and linear polarization resistance measurements, cyclic polarization measurements, and electrochemical impedance spectroscopy in 9 g L−1 NaCl, show that TiNi has satisfactory corrosion stability. According to the SEM and EDS analysis, after cyclic polarization, pitting corrosion occurred, accompanied by the dissolution of the unstable Ti2Ni inclusions. The analysis also showed that TiNi has good biocompatibility for human osteoblast-like cells, as determined by the mitochondrial activity, which was assessed using a direct contact test following ISO standard 10993-5, via scanning electron microscopy (SEM) and fluorescent microscopy. Full article
(This article belongs to the Special Issue Advances in Lightweight Alloys, 2nd Edition)
Show Figures

Figure 1

13 pages, 2115 KiB  
Article
Residual-Free Micro–Nano Titanium Surfaces via Titanium Blasting and Single Acid-Etching: A Cleaner Alternative
by Artiom Lijnev, José Eduardo Maté Sánchez de Val, Jeevithan Elango, Carlos Pérez-Albacete Martínez, José Manuel Granero Marín, Antonio Scarano and Sergio Alexandre Gehrke
Bioengineering 2025, 12(7), 735; https://doi.org/10.3390/bioengineering12070735 - 5 Jul 2025
Viewed by 648
Abstract
Background: Traditional sandblasted large-grit acid-etched (SLA) surface treatments frequently utilize alumina (Al2O3) blasting, which may leave residual particles embedded in implant surfaces, potentially compromising biocompatibility and osseointegration. This study investigates a contamination-free alternative: titanium dioxide particle (TiO2) [...] Read more.
Background: Traditional sandblasted large-grit acid-etched (SLA) surface treatments frequently utilize alumina (Al2O3) blasting, which may leave residual particles embedded in implant surfaces, potentially compromising biocompatibility and osseointegration. This study investigates a contamination-free alternative: titanium dioxide particle (TiO2) blasting followed by hydrochloric acid (HCl) etching, aimed at generating a cleaner, hierarchical micro–nano-textured surface. Methods: Grade IV titanium disks were treated either with TiO2 sandblasting alone or with an additional HCl etching step. Surfaces were analyzed via atomic force microscopy (AFM), scanning electron microscopy (SEM), contact angle measurements, and profilometry. hFOB osteoblasts were cultured to assess adhesion, proliferation, metabolic activity, and morphology. Results: The combination treatment produced a more homogeneous micro–nano structure with significantly increased roughness and a cleaner surface chemistry. Osteoblast proliferation and metabolic activity were notably improved in the TiO2 and HCl group. SEM imaging showed a more organized cytoskeletal structure and pronounced filopodia at 72 h. Conclusions: Titanium blasting combined with HCl etching yields a cost-effective, contamination-free surface modification with promising early-stage cellular responses. This approach represents a safer and effective alternative to conventional SLA treatment. Full article
(This article belongs to the Special Issue Periodontics and Implant Dentistry)
Show Figures

Figure 1

16 pages, 3074 KiB  
Review
The Role of Autophagy in the Mineralization Process of Bone and Dentin
by Ian Moran, Cassandra Villani and Anne George
Int. J. Mol. Sci. 2025, 26(13), 6278; https://doi.org/10.3390/ijms26136278 - 29 Jun 2025
Viewed by 543
Abstract
Autophagy is a cellular process that recycles intracellular macromolecules and degrades toxic cytoplasmic material to provide the cell with nutrients and facilitate survival. Although autophagy and its role in the differentiation of osteoblasts, osteoclasts, and odontoblasts has been described, the importance of autophagy [...] Read more.
Autophagy is a cellular process that recycles intracellular macromolecules and degrades toxic cytoplasmic material to provide the cell with nutrients and facilitate survival. Although autophagy and its role in the differentiation of osteoblasts, osteoclasts, and odontoblasts has been described, the importance of autophagy during matrix mineralization remains unaddressed. This review aims to characterize the autophagy/matrix mineralization relationship and elucidate the significance of autophagy during matrix mineralization. During the mineralization process, autophagy is important for cell survival and promotes the differentiation of osteoblasts and odontoblasts, the key cells that facilitate bone and dentin formation. Differentiation of these cells results in the synthesis of an organic proteinaceous matrix which subsequently forms the template for the deposition of calcium and phosphate to ultimately form crystalline hydroxyapatite. In bone, autophagy influences osteoblastic/osteoclastic activity and bone remodeling. In dentin, autophagy participates in odontogenic differentiation and facilitates odontoblastic secretion of dentin matrix proteins. This review aims to show that autophagy is critical for bone mineralization and tooth formation by supporting intracellular signaling pathways required for cell differentiation and subsequent matrix mineralization. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Back to TopTop