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
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
remove_circle_outline
remove_circle_outline

Search Results (1,746)

Search Parameters:
Keywords = bone repairing

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 2956 KiB  
Article
Impact of Photobiomodulation on the Pro-Osteogenic Activity of Dental Pulp Mesenchymal Stem/Stromal Cells
by Marcella Rodrigues Ueda Fernandes, Gabriella Teti, Valentina Gatta, Aurora Longhin, Ana Cecilia Corrêa Aranha and Mirella Falconi
Int. J. Mol. Sci. 2025, 26(17), 8174; https://doi.org/10.3390/ijms26178174 - 22 Aug 2025
Abstract
Photobiomodulation (PBM) consists of applying low-level laser light to biological tissues, leading to modulation of cellular functions. PBM has recently gained much attention in the field of regenerative dentistry thanks to its powerful effect on tissue repair and regeneration. Dental pulp mesenchymal stem/stromal [...] Read more.
Photobiomodulation (PBM) consists of applying low-level laser light to biological tissues, leading to modulation of cellular functions. PBM has recently gained much attention in the field of regenerative dentistry thanks to its powerful effect on tissue repair and regeneration. Dental pulp mesenchymal stem/stromal cells (DP-MSCs) represent the ideal targets in regenerative dentistry due to their ability to stimulate the regeneration of mineralized and soft tissues and the paracrine factors that they produce. Although there have been several studies evaluating the influence of PBM on DP-MSCs’ regenerative capacity, the results are conflicting, and there are few studies on the influence of PBM on the paracrine factors released by DP-MSCs. Therefore, the aim of this study was to investigate the effect of PBM, using different energy doses of laser irradiation, on the osteogenic capacity of DP-MSCs, focusing on changes in gene expression, mineralizing ability, and release of pro-osteogenic factors. DP-MSCs were irradiated in vitro and differentiated into an osteogenic phenotype. A cell viability assay, alizarin red staining, and TEM analysis were carried out to evaluate the effect of PBM on cell activity, morphology, and mineralization ability. The expression of the main osteogenesis-related markers Runx2, Col1A1, ALP, and BMP was measured to evaluate the influence of PBM on the ability of DP-MSCs to differentiate toward an osteogenic phenotype. The release of IL-6 and IL-8, which are mainly involved in bone remodeling processes, was investigated in the cell medium following PBM irradiation. The results showed a high level of cell viability, suggesting a lack of phototoxicity under the tested conditions. Furthermore, PBM had a significant effect on mineral deposition, IL-6 and IL-8 release, and expression of osteogenic markers. TEM analysis showed intracellular modifications linked mainly to mitochondria, the endoplasmic reticulum, and autophagic vesicles after PBM treatment. These findings demonstrated that the impact of PBM on the osteogenic potential of DP-MSCs is energy dose-dependent, supporting its potential as an effective strategy in regenerative dentistry, particularly for enhancing bone remodeling. Full article
(This article belongs to the Special Issue Application of Biotechnology to Dental Treatment)
Show Figures

Figure 1

9 pages, 2221 KiB  
Case Report
Orbital Cellutitis and Peri-Zygomatic Cutaneous Fistula After Monolateral Double Zygomatic Implant Placement: Case Report and Narrative Literature Review
by Domenico Sfondrini, Stefano Marelli, Dario De Martis, Andrea Scribante, Giada Beltramini, Luca Autelitano and Lorenzo Preda
Dent. J. 2025, 13(8), 381; https://doi.org/10.3390/dj13080381 - 21 Aug 2025
Abstract
Background. The use of zygomatic implants (ZIs) provides a highly predictable treatment option for rehabilitation in patients with severe atrophic maxillae. However, these long implants can potentially cause a number of more serious complications than those seen with conventional dental implants. The aim [...] Read more.
Background. The use of zygomatic implants (ZIs) provides a highly predictable treatment option for rehabilitation in patients with severe atrophic maxillae. However, these long implants can potentially cause a number of more serious complications than those seen with conventional dental implants. The aim of this study is to report a case of peri-zygomatic cutaneous fistula following placement of monolateral double zygomatic implants and to analyse the available literature on this complication. Methods. The 55-year-old patient was treated with placement of 3 ZIs, two on the left side. Left periorbital swelling with pain appeared 10 days after surgery with progressive worsening of symptoms. After antibiotic treatment, she developed a left cutaneous fistula with purulent discharge. CT showed two ZIs on the left side with the apical portions in close contact with a 1 cm-wide portion of resorbed zygomatic external cortex and a layer of granulation tissue. Results: Due to the limited amount of bone involved by the fixation tip, the left ZIs were removed and the skin fistula repaired. The patient healed without complications but required prosthesis replacement. Conclusions. After conducting a literature review, peri-zygomatic fistulas seem to be more common in patients with two ZIs placed on the same zygoma. In this case, the amount of available zygomatic bone is relatively limited; the bone drill holes can often be too close together and cause overheating, leading to inter-implant bone resorption and infection, with further orbito-zygomatic fistula development. The authors identified the lack of distance between ZI fixtures as one of the main causes of extraoral ZI infection. Full article
(This article belongs to the Special Issue Oral Implantology and Rehabilitation)
Show Figures

Figure 1

23 pages, 1615 KiB  
Review
Current Mechanobiological Pathways and Therapies Driving Spinal Health
by Rahul Kumar, Kyle Sporn, Harlene Kaur, Akshay Khanna, Phani Paladugu, Nasif Zaman and Alireza Tavakkoli
Bioengineering 2025, 12(8), 886; https://doi.org/10.3390/bioengineering12080886 - 20 Aug 2025
Viewed by 217
Abstract
Spinal health depends on the dynamic interplay between mechanical forces, biochemical signaling, and cellular behavior. This review explores how key molecular pathways, including integrin, yeas-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), Piezo, and Wingless/Integrated (Wnt) with β-catenin, actively shape the [...] Read more.
Spinal health depends on the dynamic interplay between mechanical forces, biochemical signaling, and cellular behavior. This review explores how key molecular pathways, including integrin, yeas-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), Piezo, and Wingless/Integrated (Wnt) with β-catenin, actively shape the structural and functional integrity of spinal tissues. These signaling mechanisms respond to physical cues and interact with inflammatory mediators such as interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor alpha (TNF-α), driving changes that lead to disc degeneration, vertebral fractures, spinal cord injury, and ligament failure. New research is emerging that shows scaffold designs that can directly harness these pathways. Further, new stem cell-based therapies have been shown to promote disc regeneration through targeted differentiation and paracrine signaling. Interestingly, many novel bone and ligament scaffolds are modulating anti-inflammatory signals to enhance tissue repair and integration, as well as prevent scaffold degradation. Neural scaffolds are also arising. These mimic spinal biomechanics and activate Piezo signaling to guide axonal growth and restore motor function. Scientists have begun combining these biological platforms with brain–computer interface technology to restore movement and sensory feedback in patients with severe spinal damage. Although this technology is not fully clinically ready, this field is advancing rapidly. As implantable technology can now mimic physiological processes, molecular signaling, biomechanical design, and neurotechnology opens new possibilities for restoring spinal function and improving the quality of life for individuals with spinal disorders. Full article
(This article belongs to the Special Issue Biomechanics and Mechanobiology in Cell and Tissue Engineering)
Show Figures

Figure 1

18 pages, 6192 KiB  
Article
Titanium Implants Functionalized with Zoledronic Acid Associated with Ruterpy Accelerate Peri-Implant Repair in Healthy and Osteoporotic Rats
by Laura Vidoto Paludetto, Isadora Breseghello, Sabrina Cruz Tfaile Frasnelli, Fábio Roberto de Souza Batista, Paulo Roberto Botacin, Cristina Antoniali, Paulo Noronha Lisboa-Filho and Roberta Okamoto
Biomimetics 2025, 10(8), 547; https://doi.org/10.3390/biomimetics10080547 - 20 Aug 2025
Viewed by 143
Abstract
Osteoporosis compromises bone quality and impairs implant osseointegration. Since an adequate bone bed is essential for implant stability and success, this study evaluated the effects of implant surface functionalization with zoledronic acid (ZOL), alone or combined with ruterpy (TERPY), on peri-implant bone healing [...] Read more.
Osteoporosis compromises bone quality and impairs implant osseointegration. Since an adequate bone bed is essential for implant stability and success, this study evaluated the effects of implant surface functionalization with zoledronic acid (ZOL), alone or combined with ruterpy (TERPY), on peri-implant bone healing in healthy (SHAM) and osteoporotic (OVX) rats. ZOL has antiresorptive properties, while TERPY exhibits osteoinductive potential. The hypothesis was that ZOL + TERPY would act synergistically by inhibiting bone resorption and promoting new bone formation. Sixty-six female Wistar rats (3 months old) were divided into six groups (n = 11) according to systemic condition (SHAM or OVX) and implant type: conventional (CONV), ZOL, or ZOL + TERPY. Surgeries (sham or bilateral ovariectomy) were performed on day 0, and implants were placed in the tibial metaphysis on day 90. Fluorochromes were administered on days 104 (calcein) and 114 (alizarin), and euthanasia was performed on day 118. Samples were analyzed histologically via confocal microscopy and micro-computed tomography (Micro-CT). The ZOL + TERPY groups demonstrated significantly accelerated peri-implant bone repair, showing greater bone formation and organization; improved BV/TV, Tb.N, and I.S.; and reduced Tb.Sp and Po.Tot compared to CONV and ZOL-alone groups. In conclusion, ZOL + TERPY enhances and speeds bone healing, even under osteoporotic conditions. Full article
Show Figures

Figure 1

21 pages, 6478 KiB  
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 379
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

14 pages, 4450 KiB  
Article
Photothermally Responsive Biomimetic Composite Scaffolds Based on Polydopamine-Functionalized Nanoparticles/Polyurethane for Bone Repair
by Ruqing Bai, Jiaqi Chen, Ting Zhang, Tao Chen, Xiaoying Liu, Weihu Yang, Tuck-Whye Wong, Jianwei Zhang and Li Wang
J. Funct. Biomater. 2025, 16(8), 294; https://doi.org/10.3390/jfb16080294 - 15 Aug 2025
Viewed by 461
Abstract
In this study, a shape-changeable 3D scaffold with photothermal effects was developed to address the clinical challenges of complex bone defects. The multifunctional construct was fabricated via in situ polymerization combined with a gas foaming technique, creating hierarchical porous architectures that mimic the [...] Read more.
In this study, a shape-changeable 3D scaffold with photothermal effects was developed to address the clinical challenges of complex bone defects. The multifunctional construct was fabricated via in situ polymerization combined with a gas foaming technique, creating hierarchical porous architectures that mimic the native bone extracellular matrix. By incorporating polydopamine (PDA)-modified amorphous calcium phosphate (CA) into poly(propylene glycol) (PPG)- and poly(ԑ-caprolactone) (PCL)-based polyurethane (PU). The obtained scaffolds achieved osteoinductive potential for bone tissue engineering. The surface PDA modification of CA enabled efficient photothermal shape conversion under near-infrared (NIR) irradiation, facilitating non-invasive remote control of localized hyperthermia. The optimized scaffolds exhibited interconnected porosity (approximately 70%) with osteoconductive pore channels (200–500 μm), resulting in good osteoinduction in cell culture, and precise shape-memory recovery at physiological temperatures (~40 °C) under NIR for minimally invasive delivery. The synergistic effect of osteogenesis promotion and photothermal transition demonstrated this programmable scaffold as a promising solution for integrated minimally invasive bone repair and defect reconstruction. Full article
Show Figures

Figure 1

20 pages, 3892 KiB  
Article
Necrotic Bone Fluid Suppresses Energy Metabolism of Porcine PBMC-Derived Macrophages In Vitro
by Zhuo Deng, Chau P. Nguyen, Yan Liu, Jaehyup Kim, Thomas P. Mathews, Chi Ma, Yinshi Ren, Chao Xing and Harry K. W. Kim
Cells 2025, 14(16), 1258; https://doi.org/10.3390/cells14161258 - 14 Aug 2025
Viewed by 217
Abstract
Legg–Calvé–Perthes disease is a juvenile ischemic osteonecrosis (ON) of the femoral head. A disruption of blood supply to the femoral head produces extensive cell death and necrotic debris. Macrophages are innate immune cells recruited to the necrotic bone to orchestrate the repair process. [...] Read more.
Legg–Calvé–Perthes disease is a juvenile ischemic osteonecrosis (ON) of the femoral head. A disruption of blood supply to the femoral head produces extensive cell death and necrotic debris. Macrophages are innate immune cells recruited to the necrotic bone to orchestrate the repair process. However, the role macrophages play in the ON repair process is still not elucidated. The purpose of this study was to determine the effect of artificial necrotic bone fluid (NBF) on porcine peripheral blood mononuclear cell (PBMC)-derived macrophages. Monocytes were positively selected by CD14 MicroBeads from pig PBMCs. After maturation, cells were treated with no stimulant (Con), LPS + IFNγ (M1), IL4 + IL13 (M2), or NBF. All culture supernatants and cells were harvested for ELISA, Western blot, FACS, RT-qPCR and bulk RNAseq. The Western blot and ELISA showed that only the M1 condition elevated the protein level of pro-inflammatory cytokines. The FACS results indicated that percentage of CD8086+ (M1 marker) cells was significantly lower in the M2 vs. other conditions, whereas the relative median fluorescence intensity of CD8086 was significantly higher in the M1 vs. other conditions. The NBF did not show any significant change compared to the Con. mRNA analysis showed significantly increased IL1β and IL8 expression in the M1 vs. Con scenario. TNFα expression was significantly decreased in the M2 vs. Con scenario. Interestingly, the NBF did not induce pro-inflammatory gene expression. For bulk RNAseq, the Gene Set Enrichment Analyses of the M1-stimulated cells revealed the enrichment of pro-inflammatory gene sets. For the M2, most of the enriched categories were related to the down-regulation of inflammation. For the NBF, the most enriched categories were related to the down-regulation of protein translation and mitochondrial metabolism. We further confirmed the suppressive effects of NBF on macrophage functions using Seahorse Cell Mito Stress Tests, 13C-glucose metabolic flux analysis, mitochondrial ROS detection via MitoSOXTM staining, and phagocytosis assay. Taken together, these results revealed that the artificial NBF down-regulates the overall cellular activity and energy metabolism of macrophages. Full article
Show Figures

Figure 1

15 pages, 3491 KiB  
Article
PARP Inhibition Shifts Murine Myeloid Cells Toward a More Tolerogenic Profile In Vivo
by Jose R. Pittaluga-Villarreal, Casey M. Daniels, Tara Capece, Pauline R. Kaplan, Martin Meier-Schellersheim and Aleksandra Nita-Lazar
Biomolecules 2025, 15(8), 1149; https://doi.org/10.3390/biom15081149 - 9 Aug 2025
Viewed by 446
Abstract
The human Poly ADP-ribose Polymerase (PARP) family comprises 17 enzymes responsible for the transfer of ADP-ribose to proteins, forming poly- or mono-ADP-ribosylation. This post-translational modification regulates DNA repair and programmed cell death, processes affecting cancer biology. PARP inhibitors, including the FDA-approved olaparib, are [...] Read more.
The human Poly ADP-ribose Polymerase (PARP) family comprises 17 enzymes responsible for the transfer of ADP-ribose to proteins, forming poly- or mono-ADP-ribosylation. This post-translational modification regulates DNA repair and programmed cell death, processes affecting cancer biology. PARP inhibitors, including the FDA-approved olaparib, are used to treat BRCA-dependent breast and ovarian cancers. Although therapies with use of PARP inhibitors are showing clinical success, their effects on the immune system remain understudied. Prior work has shown that PARP inhibition can modulate inflammatory responses and alter innate immunity. In this study, we evaluated the immunomodulatory effects of olaparib on myeloid cells in vivo, focusing on bone marrow and spleen. Olaparib treatment altered the composition and activation state of dendritic cells, neutrophils, and macrophages. In the bone marrow, olaparib increased the proportion of cDC2 population, mature neutrophils and inflammatory macrophages expressing CD80. In contrast, splenic myeloid cells exhibited enhanced expression of markers associated with tolerogenic phenotypes, including CD206 and CD124 in neutrophils and macrophages. The spleen also showed an increase in immature monocyte-derived dendritic cells (CD206+) and a bias toward the cDC2 subset. These findings indicate that PARP inhibition can induce short-term phenotypic remodeling of myeloid cell populations, promoting a more immunoregulatory profile, especially in the spleen. These changes may contribute to an altered immune landscape with implications for anti-tumor immunity. Full article
(This article belongs to the Special Issue PARPs in Cell Death and PARP Inhibitors in Cancers: 2nd Edition)
Show Figures

Figure 1

25 pages, 4674 KiB  
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 589
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

26 pages, 769 KiB  
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 644
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

25 pages, 3822 KiB  
Article
Comparative Transcriptome and MicroRNA Profiles of Equine Mesenchymal Stem Cells, Fibroblasts, and Their Extracellular Vesicles
by Sebastian Sawicki, Monika Bugno-Poniewierska, Jakub Żurowski, Tomasz Szmatoła, Ewelina Semik-Gurgul, Michał Bochenek, Elżbieta Karnas and Artur Gurgul
Genes 2025, 16(8), 936; https://doi.org/10.3390/genes16080936 - 5 Aug 2025
Viewed by 848
Abstract
Background: Mesenchymal stem cells (MSCs) are a promising tool in regenerative medicine due to their ability to secrete paracrine factors that modulate tissue repair. Extracellular vesicles (EVs) released by MSCs contain bioactive molecules (e.g., mRNAs, miRNAs, proteins) and play a key role in [...] Read more.
Background: Mesenchymal stem cells (MSCs) are a promising tool in regenerative medicine due to their ability to secrete paracrine factors that modulate tissue repair. Extracellular vesicles (EVs) released by MSCs contain bioactive molecules (e.g., mRNAs, miRNAs, proteins) and play a key role in intercellular communication. Methods: This study compared the transcriptomic profiles (mRNA and miRNA) of equine MSCs derived from adipose tissue (AT-MSCs), bone marrow (BM-MSCs), and ovarian fibroblasts (as a differentiated control). Additionally, miRNAs present in EVs secreted by these cells were characterized using next-generation sequencing. Results: All cell types met ISCT criteria for MSCs, including CD90 expression, lack of MHC II, trilineage differentiation, and adherence. EVs were isolated using ultracentrifugation and validated with nanoparticle tracking analysis and flow cytometry (CD63, CD81). Differential expression analysis revealed distinct mRNA and miRNA profiles across cell types and their secreted EVs, correlating with tissue origin. BM-MSCs showed unique regulation of genes linked to early development and osteogenesis. EVs contained diverse RNA species, including miRNA, mRNA, lncRNA, rRNA, and others. In total, 227 and 256 mature miRNAs were detected in BM-MSCs and AT-MSCs, respectively, including two novel miRNAs per MSC type. Fibroblasts expressed 209 mature miRNAs, including one novel miRNA also found in MSCs. Compared to fibroblasts, 60 and 92 differentially expressed miRNAs were identified in AT-MSCs and BM-MSCs, respectively. Conclusions: The results indicate that MSC tissue origin influences both transcriptomic profiles and EV miRNA content, which may help to interpret their therapeutic potential. Identifying key mRNAs and miRNAs could aid in future optimizing of MSC-based therapies in horses. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

19 pages, 94974 KiB  
Article
Promotion of Bone Defect Repair Using Decellularized Antler Cancellous Bone Loaded with Deer Osteoglycin
by Yusu Wang, Ying Zong, Weijia Chen, Naichao Diao, Quanmin Zhao, Boyin Jia, Miao Zhang, Jianming Li, Yan Zhao, Zhongmei He and Rui Du
Biomolecules 2025, 15(8), 1124; https://doi.org/10.3390/biom15081124 - 4 Aug 2025
Viewed by 388
Abstract
The combination of scaffold materials and bioactive factors is a promising strategy for promoting bone defect repair in tissue engineering. Previous studies have shown that osteoglycin (OGN) is highly expressed in the bone repair process using deer antler as an animal model of [...] Read more.
The combination of scaffold materials and bioactive factors is a promising strategy for promoting bone defect repair in tissue engineering. Previous studies have shown that osteoglycin (OGN) is highly expressed in the bone repair process using deer antler as an animal model of bone defects. It suggests that OGN may be a key active component involved in the bone repair process. The aim of this study was to investigate whether deer OGN (dOGN) could effectively promote bone regeneration. We successfully expressed dOGN using the E. coli pET30a system and evaluated its biological activity through cell proliferation and migration assays. At a concentration of 5 μg/mL, dOGN significantly promoted cell proliferation and migration. We then incorporated dOGN onto decellularized antler cancellous bone (DACB) scaffolds and assessed their osteogenic potential both in vitro and in vivo. The results indicated that dOGN loading enhanced cell proliferation, adhesion, and osteogenic activity. In vivo experiments confirmed that the dOGN-DACB scaffold significantly improved bone regeneration compared to DACB alone. This study demonstrates that dOGN-loaded DACB scaffolds hold great potential for clinical applications in treating critical-sized bone defects by mimicking the rapid regenerative properties of deer antlers. Full article
(This article belongs to the Special Issue Tissue Calcification in Normal and Pathological Environments)
Show Figures

Figure 1

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 783
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

22 pages, 1937 KiB  
Review
Carbon Dot Nanozymes in Orthopedic Disease Treatment: Comprehensive Overview, Perspectives and Challenges
by Huihui Wang
C 2025, 11(3), 58; https://doi.org/10.3390/c11030058 - 1 Aug 2025
Viewed by 410
Abstract
Nanozymes, as a new generation of artificial enzymes, have attracted increasing attention in the field of biomedicine due to their multiple enzymatic characteristics, multi-functionality, low cost, and high stability. Among them, carbon dot nanozymes (CDzymes) possess excellent enzymatic-like catalytic activity and biocompatibility and [...] Read more.
Nanozymes, as a new generation of artificial enzymes, have attracted increasing attention in the field of biomedicine due to their multiple enzymatic characteristics, multi-functionality, low cost, and high stability. Among them, carbon dot nanozymes (CDzymes) possess excellent enzymatic-like catalytic activity and biocompatibility and have been developed for various diagnostic and therapeutic studies of diseases. Here, we briefly review the representative research on CDzymes in recent years, including their synthesis, modification, and applications, especially in orthopedic diseases, including osteoarthritis, osteoporosis, osteomyelitis, intervertebral disc degenerative diseases, bone tumors, and bone injury repair and periodontitis. Additionally, we briefly discuss the potential future applications and opportunities and challenges of CDzymes. We hope this review can provide some reference opinions for CDzymes and offer insights for promoting their application strategies in the treatment of orthopedic disease. Full article
(This article belongs to the Special Issue Carbon Nanohybrids for Biomedical Applications (2nd Edition))
Show Figures

Figure 1

15 pages, 2399 KiB  
Review
Cyclodextrin-Based Supramolecular Hydrogels in Tissue Engineering and Regenerative Medicine
by Jiamin Lin, Yuanyuan Chen and Xuemei Wang
Molecules 2025, 30(15), 3225; https://doi.org/10.3390/molecules30153225 - 31 Jul 2025
Viewed by 498
Abstract
Cyclodextrins (CDs), cyclic oligosaccharides formed by α-1,4-glycosidic-bonded D-glucopyranose units, feature unique hydrophobic cavities and hydrophilic exteriors that enable molecular encapsulation via host–guest interactions. CDs form supramolecular host–guest complexes with diverse molecular entities, establishing their fundamental role in supramolecular chemistry. This review examines fabrication [...] Read more.
Cyclodextrins (CDs), cyclic oligosaccharides formed by α-1,4-glycosidic-bonded D-glucopyranose units, feature unique hydrophobic cavities and hydrophilic exteriors that enable molecular encapsulation via host–guest interactions. CDs form supramolecular host–guest complexes with diverse molecular entities, establishing their fundamental role in supramolecular chemistry. This review examines fabrication strategies for CD-based supramolecular hydrogels and their applications in tissue engineering and regenerative medicine, with focused analysis on wound healing, corneal regeneration, and bone repair. We critically analyze CD–guest molecular interaction mechanisms and innovative therapeutic implementations, highlighting the significant potential of CD hydrogels for tissue regeneration while addressing clinical translation challenges and future directions. Full article
(This article belongs to the Special Issue Cyclodextrin Chemistry and Toxicology III)
Show Figures

Figure 1

Back to TopTop