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
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
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 (465)

Search Parameters:
Keywords = bone therapeutic agents

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 3608 KB  
Article
Osteogenic-Inducing Apatite/Agarose/Gelatin Hybrid Scaffolds Embedding Gold Nanoparticles
by María Victoria Cabañas, Paola S. Padilla, Mónica Cicuéndez, Sandra Sánchez-Salcedo, Jesús Román and Juan Peña
Pharmaceutics 2025, 17(9), 1103; https://doi.org/10.3390/pharmaceutics17091103 - 25 Aug 2025
Abstract
Objectives: To prepare porous scaffolds combining hydrogel and hydroxycarbonateapatite, enriched with a promising therapeutic agent, gold nanoparticles, to improve bone regeneration. The fabrication procedure is conducted under mild conditions, without toxic or aggressive chemicals, at physiological pH, and low temperatures; Methods: [...] Read more.
Objectives: To prepare porous scaffolds combining hydrogel and hydroxycarbonateapatite, enriched with a promising therapeutic agent, gold nanoparticles, to improve bone regeneration. The fabrication procedure is conducted under mild conditions, without toxic or aggressive chemicals, at physiological pH, and low temperatures; Methods: Gold nanoparticles (15–20 nm), were obtained by the Turkevith method. The scaffolds were fabricated by the GELPOR3D method, which has demonstrated its ability to integrate thermal labile molecules, during the scaffold fabrication process. The role of these nanoparticles in promoting cell adhesion, proliferation, and mineralization processes in vitro has been studied using osteoprogenitor MC3T3-E1 cells; Results: The scaffold fabrication conditions, combined with the surface functionalization of the gold nanoparticles with poly(ethylene glycol), ensure their uniform distribution throughout the scaffold and facilitate their gradual release over 48 h in a physiological medium. A significant increase in the mean cell area and a significant decrease in the circularity index during the early stages of osteoblast differentiation are observed. These pieces of evidence suggest that adequate cell spreading could lead to enhanced proliferation and matrix deposition activity; Conclusions: Scaffolds containing these gold nanoparticles exhibited a marked improvement in adhesion, proliferation, and mineralization of preosteoblasts (MC3T3 cells) at the concentrations studied. The functionalization of the nanoparticles, along with the shaping procedure employed, is critical for their homogeneous dispersion throughout the scaffold and their progressive release. The findings confirm the crucial role of gold nanoparticles in the early stages of osteoblast differentiation, which is essential for the transition from premature osteoblasts to mature osteoblasts. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Figure 1

14 pages, 11268 KB  
Article
Hyaluronan-Related Granulomatous Synovitis, Adipositis, and Osteomyelitis in the Osteoarthritic Knee: A Morphological Case Series of Three Patients
by Vera Lyalina, Gulnara Eshmotova, Alexandra Karavan, Andrey Korshunov, Alexey Zarov, Anton Bonartsev, Natalia Serejnikova, Alexey Prizov, George Airapetov and Alexey Volkov
Int. J. Mol. Sci. 2025, 26(16), 8073; https://doi.org/10.3390/ijms26168073 - 21 Aug 2025
Viewed by 149
Abstract
Intra-articular hyaluronan injections represent a widely used and generally safe therapeutic approach for knee osteoarthritis (OA). However, the side effects of this treatment remain insufficiently studied. Acute post-injection reactions, particularly those arising from an improper technique resulting in the deposition of the therapeutic [...] Read more.
Intra-articular hyaluronan injections represent a widely used and generally safe therapeutic approach for knee osteoarthritis (OA). However, the side effects of this treatment remain insufficiently studied. Acute post-injection reactions, particularly those arising from an improper technique resulting in the deposition of the therapeutic agent into joint tissues, are well-documented. In contrast, chronic hyaluronan-induced inflammatory responses have received scant attention in the scientific literature. The aim of this study is to characterize for the first time the morphological patterns of chronic granulomatous inflammation induced by exogenous hyaluronan (e-HA) in osteoarthritic knees, focusing on three distinct tissue reactions: synovitis, adipositis, and osteomyelitis. Using a three-case series approach and morphological analysis, we identified e-HA penetration pathways; described associated foreign body responses in the synovial, adipose, and bone tissues of the joints; and emphasized the clinical relevance of these underreported adverse effects. These observations highlight an understudied phenomenon—an active conflict between e-HA and joint tissues that recognize it as a foreign body. The prevalence, clinical significance, and prognostic implications of this phenomenon require further investigation. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapeutic Approaches to Osteoarthritis)
Show Figures

Figure 1

9 pages, 934 KB  
Case Report
Pediatric Acute Megakaryoblastic Leukemia with a GATA2 Mutation and Monosomy 7: A Case Report and Clinical Management Challenges
by Gowri Joshi, Astil Jisho Anto, Md Maaz Mallick, Gwan Yong Lim and Łukasz Hutnik
Reports 2025, 8(3), 153; https://doi.org/10.3390/reports8030153 - 21 Aug 2025
Viewed by 214
Abstract
Background and Clinical Significance: Acute megakaryoblastic leukemia (AMKL) is a rare and aggressive hematologic malignancy. The presence of genetic abnormalities often increases the complexity of AMKL. Among these, patients with monosomy 7 constitute a high-risk group associated with a poorer prognosis and [...] Read more.
Background and Clinical Significance: Acute megakaryoblastic leukemia (AMKL) is a rare and aggressive hematologic malignancy. The presence of genetic abnormalities often increases the complexity of AMKL. Among these, patients with monosomy 7 constitute a high-risk group associated with a poorer prognosis and greater chemoresistance. We report the case of a 10-year-old boy who had AMKL along with monosomy 7 and familial GATA2 deficiency. The case highlights the diagnostic and therapeutic challenges faced, as well as the critical importance of early genetic screening and timely hematopoietic stem cell transplantation (HSCT). Case Presentation: A 10-year-old boy presented with easy bruising and pancytopenia. AMKL was diagnosed with the help of a bone marrow biopsy and immunophenotyping. Genetic testing showed a GATA2 mutation and monosomy 7. Two induction cycles with daunorubicin and cytarabine were administered but failed to eliminate residual disease. The patient also developed pneumonia of a fungal origin. HSCT was delayed due to liver toxicity and elevated minimal residual disease (MRD). Azacitidine and venetoclax stabilized the disease, thereby allowing for successful haploidentical HSCT. The patient achieved complete remission with full donor chimerism. Conclusions: This case emphasizes the importance of early molecular diagnostics in pediatric AMKL. Identifying GATA2 mutations and monosomy 7 early can help guide risk stratification and the timing of HSCT. Multimodal therapy, which includes the use of infection control and targeted agents, is important for improving the outcomes in high-risk patients. Full article
(This article belongs to the Section Haematology)
Show Figures

Figure 1

19 pages, 1400 KB  
Systematic Review
Targeting Macrophages in Glioblastoma: Current Therapies and Future Directions
by Giovanni Pennisi, Federico Valeri, Benedetta Burattini, Placido Bruzzaniti, Carmelo Lucio Sturiale, Andrea Talacchi, Fabio Papacci, Alessandro Olivi and Giuseppe Maria Della Pepa
Cancers 2025, 17(16), 2687; https://doi.org/10.3390/cancers17162687 - 18 Aug 2025
Viewed by 418
Abstract
Glioblastoma (GBM) is an aggressive brain tumor characterized by an immunosuppressive tumor microenvironment (TME), which contributes to treatment resistance and disease progression. Background: Tumor-associated macrophages (TAMs), comprising both resident microglia and bone marrow–derived macrophages, play a central role in supporting tumor growth, [...] Read more.
Glioblastoma (GBM) is an aggressive brain tumor characterized by an immunosuppressive tumor microenvironment (TME), which contributes to treatment resistance and disease progression. Background: Tumor-associated macrophages (TAMs), comprising both resident microglia and bone marrow–derived macrophages, play a central role in supporting tumor growth, angiogenesis, and immune evasion. Most TAMs adopt an M2-like immunosuppressive phenotype, making them a promising target for immunomodulatory strategies in GBM. Method: According to PRISMA guidelines, we conducted a systematic literature review and recruited eligible studies focused on therapeutic approaches targeting TAMs in GBM, emphasizing mechanisms of action, efficacy, and challenges. Data extraction focused on therapeutic classes, outcomes, and TAM-related biomarkers. Results: We identified 30 studies meeting the inclusion criteria. These therapies are categorized into three main strategies: inhibition of TAM recruitment, enhancement of TAM-mediated phagocytosis, and reprogramming of TAMs. Combination strategies, including TAM-targeting with checkpoint inhibitors, nanoparticles, and oncolytic viruses, show synergistic effects in preclinical models. Conclusions: Targeting TAMs represents a multifaceted strategy for GBM treatment. Current evidence underscores the need for combination approaches integrating TAM modulation with existing standard-of-care therapies. Clinical translation remains limited due to challenges such as TAM heterogeneity, plasticity, immunosuppressive therapies, and restricted drug delivery across the blood–brain barrier. Future directions should highlight personalized treatments based on detailed TME profiling. Combining TAM-targeted therapies with agents modulating metabolic or immune pathways, and leveraging advanced delivery systems and spatial transcriptomics may improve efficacy. Full article
Show Figures

Figure 1

21 pages, 6478 KB  
Article
Localized Combination Therapy Using Collagen–Hydroxyapatite Bone Grafts for Simultaneous Bone Cancer Inhibition and Tissue Regeneration
by Alina Florentina Vladu, Madalina Georgiana Albu Kaya, Anton Ficai, Denisa Ficai, Raluca Tutuianu, Ludmila Motelica, Vasile Adrian Surdu, Ovidiu-Cristian Oprea, Roxana Doina Truşcă and Irina Titorencu
Polymers 2025, 17(16), 2239; https://doi.org/10.3390/polym17162239 - 18 Aug 2025
Viewed by 435
Abstract
The global burden of cancer continues to grow, with bone cancer—though rare—posing serious challenges in terms of treatment and post-surgical reconstruction. Autologous bone grafting remains the gold standard, yet limitations such as donor site morbidity drive the search for alternative solutions. Tissue engineering, [...] Read more.
The global burden of cancer continues to grow, with bone cancer—though rare—posing serious challenges in terms of treatment and post-surgical reconstruction. Autologous bone grafting remains the gold standard, yet limitations such as donor site morbidity drive the search for alternative solutions. Tissue engineering, combining biomaterials and therapeutic agents, offers promising avenues. This study focuses on the development of multifunctional scaffolds based on collagen and hydroxyapatite obtained by the freeze-drying technique and incorporating both synthetic (doxorubicin) and natural (caffeic acid) compounds for osteosarcoma treatment. These scaffolds aim to combine tumor inhibition with bone regeneration, addressing the dual need for local drug delivery and structural repair in bone cancer therapy. The characterization of these composite materials revealed that a spongious structure with interconnected pores and a homogeneous pore distribution, with pore sizes between 20 and 250 μm suitable for osteoblasts infiltration. The Fourier transform infrared (FTIR) spectroscopy and thermogravimetric analysis-differential scanning calorimetry (TG-DSC) and X-ray diffraction (XRD) analyses confirmed the formation of hydroxyapatite inside the collagen matrix. LDH and XTT assays confirmed that the antitumoral scaffolds possess great potential for osteosarcoma treatment, showing that after 3 days of culturing, the extracts containing doxorubicin-7A, both alone and in combination with caffeic acid-9A, significantly reduced the viability of cell lines to below 7% and 20%, respectively. Full article
(This article belongs to the Special Issue Smart and Bio-Medical Polymers: 3rd Edition)
Show Figures

Figure 1

17 pages, 325 KB  
Review
The Role of Senolytics in Osteoporosis
by Erman Chen, Jingjing Zhang, Han Chen and Weixu Li
Biomolecules 2025, 15(8), 1176; https://doi.org/10.3390/biom15081176 - 16 Aug 2025
Viewed by 401
Abstract
Cellular senescence is a fundamental contributor to numerous dysfunctions and degenerative diseases, including osteoporosis. In genetically modified and preclinical animal models, therapeutic strategies targeting persistent senescent cells have been shown to delay and prevent osteoporosis. Senolytics are a class of drugs or compounds [...] Read more.
Cellular senescence is a fundamental contributor to numerous dysfunctions and degenerative diseases, including osteoporosis. In genetically modified and preclinical animal models, therapeutic strategies targeting persistent senescent cells have been shown to delay and prevent osteoporosis. Senolytics are a class of drugs or compounds designed to selectively eliminate senescent cells without adversely affecting normal cells. In this review, we focus on the role of senolytic agents in regulating bone metabolism and their potential in the treatment of osteoporosis. We discussed major types of senolytics, such as natural compounds, kinase inhibitors, Bcl-2 family inhibitors, inhibitors of the mouse double minute 2/p53 interaction, heat shock protein 90 inhibitors, p53-binding inhibitors, and histone deacetylase inhibitors. This review also highlights the progress of senolytics in clinical trials. However, clinical results diverge from preclinical evidence. Therefore, senolytics should be critically evaluated as a potential therapeutic strategy for osteoporosis, with further validation required. Full article
(This article belongs to the Special Issue Tissue Calcification in Normal and Pathological Environments)
25 pages, 4674 KB  
Review
Research Progress on Icariin Promoting Bone Injury Repair and Regeneration
by Weijian Hu, Yameng Si, Xin Xie and Jiabin Xu
Pharmaceuticals 2025, 18(8), 1174; https://doi.org/10.3390/ph18081174 - 8 Aug 2025
Viewed by 629
Abstract
Icariin (ICA) is a bioactive flavonoid compound extracted from Epimedium plants. In recent years, it has attracted significant research interest in the field of bone tissue repair due to its pharmacological effects via multiple targets and pathways. Studies have shown that ICA promotes [...] Read more.
Icariin (ICA) is a bioactive flavonoid compound extracted from Epimedium plants. In recent years, it has attracted significant research interest in the field of bone tissue repair due to its pharmacological effects via multiple targets and pathways. Studies have shown that ICA promotes the osteogenic differentiation of mesenchymal stem cells (MSCs) and enhances bone matrix formation by regulating signaling pathways such as Akt and Wnt/β-catenin. It concurrently inhibits osteoclast activity to maintain the balance of bone remodeling, thereby simultaneously stimulating new bone regeneration and suppressing bone resorption. At the same time, ICA exerts potent anti-inflammatory and antioxidant effects and promotes angiogenesis, improving the local microenvironment of bone injury and significantly facilitating the regeneration of bone and cartilage tissues. Additionally, ICA exhibits notable protective effects in multiple organ systems including the cardiovascular, hepatic, renal, and nervous systems. Specifically, ICA reduces cardiomyocyte apoptosis and fibrosis to preserve cardiac function, improves hepatic metabolic function and alleviates oxidative stress, attenuates renal inflammation and fibrosis, and—through neuroprotective actions—reduces neuroinflammation and promotes neuronal survival. These multi-organ effects help optimize the systemic environment for bone healing. However, ICA faces significant pharmacokinetic challenges. It has low oral bioavailability (due to poor absorption and extensive first-pass metabolism) as well as a short half-life. Consequently, maintaining effective drug concentrations in vivo is difficult, which limits its therapeutic efficacy and impedes clinical translation. To fully realize its regenerative potential, advanced drug delivery strategies (e.g., nanocarrier-based delivery systems) are being explored to enhance ICA’s bioavailability and prolong its duration of action. Overall, ICA’s multi-modal actions on bone cells, the immune microenvironment, and systemic factors make it a promising multi-target agent for bone regeneration. Addressing its pharmacokinetic limitations through optimized delivery and conducting further clinical studies will be crucial to realize its full therapeutic potential. This review provides a comprehensive overview of recent advances and challenges in translating ICA’s benefits into orthopedic therapy. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

30 pages, 13403 KB  
Article
Baicalein-Loaded Chitosan Films for Local Treatment of Oral Infections
by Magdalena Paczkowska-Walendowska, Anna Rył, Jakub Kwiatek, Natalia Rosiak, Kamil Szarzyński, Weronika Wawrzyniak, Julia Ziółkowska, Weronika Kuderska, Kaja Kręcka, Anna Marciniak, Tomasz M. Karpiński, Tomasz Plech, Andrzej Miklaszewski, Piotr Owczarz and Judyta Cielecka-Piontek
Polymers 2025, 17(16), 2167; https://doi.org/10.3390/polym17162167 - 8 Aug 2025
Viewed by 550
Abstract
Oral infections and tissue defects remain significant clinical challenges, often requiring localized, sustained, and multifunctional therapeutic solutions. In this study, baicalein-loaded chitosan films were developed and comprehensively characterized as novel biomaterials for oral and maxillofacial applications. Using a 32 factorial design, nine [...] Read more.
Oral infections and tissue defects remain significant clinical challenges, often requiring localized, sustained, and multifunctional therapeutic solutions. In this study, baicalein-loaded chitosan films were developed and comprehensively characterized as novel biomaterials for oral and maxillofacial applications. Using a 32 factorial design, nine film formulations were prepared via solvent casting, varying chitosan molecular weight and composition. Physicochemical and structural analyses (microscopy, SEM, FTIR, and XRPD) confirmed uniform drug distribution and matrix compatibility. Mechanical testing and dissolution studies demonstrated zero-order baicalein release kinetics, with controlled, sustained delivery influenced by chitosan content and molecular weight. The optimal formulation (F5: CS MMW 2%, Gel 2%) combined favorable mechanical integrity, drug release, and potent antioxidant and anti-inflammatory activities. Further evaluation on 3D anatomical models simulating bone and soft tissue defects highlighted excellent membrane adaptability, stability, and ease of handling under conditions mimicking clinical surgery. The films acted as effective barriers in guided tissue regeneration and donor site protection, with improved surgical visibility due to their baicalein-induced coloration. Biocompatibility assays confirmed the safety of the materials, while antibacterial testing demonstrated activity against Streptococcus mutans. These results support the potential of baicalein-loaded chitosan films as multifunctional membranes for regenerative dentistry, periodontal therapy, and peri-implant care. The modular formulation design provides a platform for future integration of additional bioactive agents, paving the way for personalized, advanced wound healing solutions. Full article
(This article belongs to the Section Biobased and Biodegradable Polymers)
Show Figures

Graphical abstract

26 pages, 769 KB  
Review
Immunomodulatory and Regenerative Functions of MSC-Derived Exosomes in Bone Repair
by Manorathna Arun, Sheeja Rajasingh, Parani Madasamy and Johnson Rajasingh
Bioengineering 2025, 12(8), 844; https://doi.org/10.3390/bioengineering12080844 - 5 Aug 2025
Viewed by 666
Abstract
Bone integrity is maintained through continuous remodeling, orchestrated by the coordinated actions of osteocytes, osteoblasts, and osteoclasts. Once considered passive bystanders, osteocytes are now recognized as central regulators of this process, mediating biochemical signaling and mechanotransduction. Malfunctioning osteocytes contribute to serious skeletal disorders [...] Read more.
Bone integrity is maintained through continuous remodeling, orchestrated by the coordinated actions of osteocytes, osteoblasts, and osteoclasts. Once considered passive bystanders, osteocytes are now recognized as central regulators of this process, mediating biochemical signaling and mechanotransduction. Malfunctioning osteocytes contribute to serious skeletal disorders such as osteoporosis. Mesenchymal stromal cells (MSCs), multipotent stem cells capable of differentiating into osteoblasts, have emerged as promising agents for bone regeneration, primarily through the paracrine effects of their secreted exosomes. MSC-derived exosomes are nanoscale vesicles enriched with proteins, lipids, and nucleic acids that promote intercellular communication, osteoblast proliferation and differentiation, and angiogenesis. Notably, they deliver osteoinductive microRNAs (miRNAs) that influence osteogenic markers and support bone tissue repair. In vivo investigations validate their capacity to enhance bone regeneration, increase bone volume, and improve biomechanical strength. Additionally, MSC-derived exosomes regulate the immune response, creating pro-osteogenic and pro-angiogenic factors, boosting their therapeutic efficacy. Due to their cell-free characteristics, MSC-derived exosomes offer benefits such as diminished immunogenicity and minimal risk of off-target effects. These properties position them as promising and innovative approaches for bone regeneration, integrating immunomodulatory effects with tissue-specific regenerative capabilities. Full article
Show Figures

Figure 1

16 pages, 948 KB  
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 270
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

25 pages, 1749 KB  
Review
TGF-β Signaling in Cancer: Mechanisms of Progression and Therapeutic Targets
by Elżbieta Cecerska-Heryć, Adrianna Jerzyk, Małgorzata Goszka, Aleksandra Polikowska, Julita Rachwalska, Natalia Serwin, Bartosz Wojciuk and Barbara Dołęgowska
Int. J. Mol. Sci. 2025, 26(15), 7326; https://doi.org/10.3390/ijms26157326 - 29 Jul 2025
Viewed by 1136
Abstract
Transforming growth factor-β (TGF-β) is a key protein family member that includes activins, inhibins, and bone morphogenetic proteins (BMPs). It is essential in numerous biological processes, such as chemotaxis, apoptosis, differentiation, growth, and cell migration. TGF-β receptors initiate signaling through two primary pathways: [...] Read more.
Transforming growth factor-β (TGF-β) is a key protein family member that includes activins, inhibins, and bone morphogenetic proteins (BMPs). It is essential in numerous biological processes, such as chemotaxis, apoptosis, differentiation, growth, and cell migration. TGF-β receptors initiate signaling through two primary pathways: the canonical pathway involving Smad proteins and non-canonical pathways that utilize alternative signaling mechanisms. When TGF-β signaling is disrupted, it has been shown to contribute to the development of various diseases, including cancer. Initially, TGF-β effectively inhibits the cell cycle and promotes apoptosis. However, its role can transition to facilitating tumor growth and metastasis as the disease progresses. Moreover, TGF-β drives cancer progression through epithelial–mesenchymal transition (EMT), modulation of factor expression, and evasion of immune responses. This complexity establishes the need for further research, particularly into pharmacological agents targeting TGF-β, which are emerging as promising therapeutic options. Current clinical and preclinical studies are making significant strides toward mitigating the adverse effects of TGF-β. This underscores the critical importance of understanding its underlying mechanisms to enhance treatment effectiveness and improve survival rates for cancer patients. Full article
(This article belongs to the Special Issue Advancements in Cancer Biomarkers)
Show Figures

Graphical abstract

31 pages, 1902 KB  
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 350
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

60 pages, 3898 KB  
Review
The Therapeutic Potential of Phytochemicals Unlocks New Avenues in the Management of Rheumatoid Arthritis
by Kalina A. Nikolova-Ganeva, Nikolina M. Mihaylova, Lidiya A. Kechidzhieva, Kristina I. Ivanova, Alexander S. Zarkov, Daniel L. Parzhanov, Momchil M. Ivanov and Andrey S. Marchev
Int. J. Mol. Sci. 2025, 26(14), 6813; https://doi.org/10.3390/ijms26146813 - 16 Jul 2025
Viewed by 911
Abstract
Rheumatoid arthritis (RA) is a progressive and systemic autoimmune disease, characterized by a chronic inflammatory process, affecting the lining of the synovial joints, many body organs/systems, and blood vessels. Its pathological hallmarks are hyperplasic synovium, bone erosion, and progressive joint destruction. Rheumatoid arthritis [...] Read more.
Rheumatoid arthritis (RA) is a progressive and systemic autoimmune disease, characterized by a chronic inflammatory process, affecting the lining of the synovial joints, many body organs/systems, and blood vessels. Its pathological hallmarks are hyperplasic synovium, bone erosion, and progressive joint destruction. Rheumatoid arthritis affects over 20 million people, with a worldwide prevalence of 0.5–1.0%, exhibiting gender, ethnic, and geographical differences. The progressive disability severely impairs physical motion and quality of life and is finally leading to a shortened life span. The pathogenesis of RA is a complex and still poorly understood process in which genetic and environmental factors are principally associated. Current treatment mostly relies on conventional/non-biological disease-modifying anti-rheumatic drugs (cDMARDs), analgesics, non-steroidal anti-inflammatory drugs, glucocorticoids, steroids, immunosuppresants, and biologic DMARDs, which only control inflammation and pain. Along with side effects (drug toxicity and intolerance), these anti-rheumatic drugs possess limited efficacy. Therefore, the discovery of novel multi-target therapeutics with an improved safety profile that function as inhibitors of RA-linked signaling systems are in high demand, and this is in the interest of both patients and clinicians. Plant-derived extracts, nutritional supplements, dietary medicine, and molecules with anti-inflammatory activity represent promising adjuvant agents or alternatives for RA therapeutics. This review not only aims to discuss the basic features of RA pathogenesis, risk factors, and signaling pathways but also highlights the research progress in pre-clinical RA in in vitro and in vivo models, revealing new avenues in the management of the disease in terms of comprehensive multidisciplinary strategies originating from medicinal plants and plant-derived molecules. Full article
(This article belongs to the Special Issue Natural Products as Multitarget Agents in Human Diseases)
Show Figures

Graphical abstract

30 pages, 3973 KB  
Review
V-ATPase and Lysosomal Energy Sensing in Periodontitis and Medicine-Related Osteonecrosis of the Jaw
by Xianrui Yang and Lexie Shannon Holliday
Biomolecules 2025, 15(7), 997; https://doi.org/10.3390/biom15070997 - 11 Jul 2025
Viewed by 557
Abstract
Diabetes is a risk factor for periodontitis. Increasing evidence suggests that a central player in this link is the vacuolar H+-ATPase (V-ATPase), which provides a physical and functional core for regulation by the catabolic lysosomal AMP-activated protein kinase complex (L-AMPK) and the anabolic [...] Read more.
Diabetes is a risk factor for periodontitis. Increasing evidence suggests that a central player in this link is the vacuolar H+-ATPase (V-ATPase), which provides a physical and functional core for regulation by the catabolic lysosomal AMP-activated protein kinase complex (L-AMPK) and the anabolic mammalian target of rapamycin complex 1 (mTORC1). These complexes detect levels of various cellular nutrients, including glucose at the lysosome, and promote cellular responses to restore homeostasis. The high-glucose conditions of diabetes foster anabolic mTORC1 signaling that increases inflammation and inflammatory bone resorption in response to periodontal infections. Here, we review the structure and composition of V-ATPase, L-AMPK, mTORC1, and other elements of the energy-sensing platform. Mechanisms by which V-ATPase passes signals to the complexes are examined and recent data are reviewed. Current anti-bone resorptive therapeutics, bisphosphonates and denosumab, enhance the risk of medicine-related osteonecrosis of the jaw (MRONJ) and are not used to treat periodontal bone loss. Accumulating data suggest that it may be possible to target inflammatory bone resorption through agents that stimulate L-AMPK, including metformin and glucagon-like peptide-1 agonists. This approach may reduce inflammatory bone resorption without major effects on overall bone remodeling or increased risk of MRONJ. Full article
Show Figures

Figure 1

29 pages, 2331 KB  
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 647
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

Back to TopTop