Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 870
Filter
1.
Acta Biomater ; 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39134130

ABSTRACT

Bone extracellular matrix (ECM) has been shown to mimic aspects of the tissue's complex microenvironment, suggesting its potential role in promoting bone repair. However, current ECM-based therapies suffer from limitations such as inefficient scale-up, lack of mechanical integrity, and sub-optimal efficacy. Here, we fabricated hydrogels from decellularized ECM (dECM) from wild type (WT) and thrombospondin-2 knockout (TSP2KO) mouse bones. TSP2KO bone ECM hydrogel was found to have distinct mechanical properties and collagen fibril assembly from WT. Furthermore, TSP2KO hydrogel promoted mesenchymal stem cell (MSC) attachment, spreading, and invasion in vitro. Similarly, it promoted formation of tube-like structures by human umbilical vein endothelial cells (HUVECs). When applied to a murine calvarial defect model, TSP2KO hydrogel enhanced repair, in part, due to increased angiogenesis. Our study suggests the pro-angiogenic therapeutic potential of TSP2KO bone ECM hydrogel in bone repair. STATEMENT OF SIGNIFICANCE: The study describes the first successful preparation of a novel hydrogel made from decellularized mouse bones. Bones from wild-type mice and mice lacking thrombospondin 2 (TSP2) were used to fabricate the gels. Hydrogels from TSP2KO bones have unique characteristics in structure and biomechanics. These gels interacted well with cells in vitro and helped repair damaged bone in a mouse model. Therefore, TSP2KO bone-derived hydrogel has translational potential for accelerating repair of bone defects that are otherwise difficult to heal. This study not only creates a new material with promise for healing, but also validates tunability of native biomaterials by genetic engineering.

2.
Int J Biol Macromol ; : 134372, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39134201

ABSTRACT

Bone tissue engineering scaffolds are an important means of repairing bone defects, but current solutions do not adequately simulate complex extracellular microenvironment fibrous structures and adjustable mechanical properties. We use template-assisted fiber freeze-shaping technology to construct silk fibroin nanofiber aerogels (SNFAs) with nanofibrous textures and adjustable mechanical properties. The parallel arranged channels, the pores, electrospun nanofibers, and silk protein conformation together constitute the hierarchical structure of SNFAs. Especially, the introduced electrospun nanofibers formed a biomimetic nanofibrous texture similar to the extracellular matrix, providing favorable conditions for cell migration and tissue regeneration. In addition, Young's modulus of SNFAs can be adjusted freely between 7 and 88 kPa. The rationally designed 3D architecture makes SNFAs perfectly mimic the fiber structure of the extracellular matrix and can adjust its mechanical properties to match the bone tissue perfectly. Finally, fiber-containing SNFAs observably promoted cell adhesion, proliferation, and differentiation, accelerating the bone repair process. The bone density in the defect area reached 0.53 g/cm3 and the bone volume/total volume (BV/TV) ratio reached 57 % at 12 weeks, respectively. It can be expected that this kind of tissue engineering scaffold with highly simulating extracellular matrix microenvironment and adjustable mechanical properties will possess broad prospects in the field of bone repair.

3.
Adv Sci (Weinh) ; : e2403201, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39137351

ABSTRACT

Angiogenesis is crucial for successful bone defect repair. Co-transplanting Bone Marrow Stromal Cells (BMSCs) and Endothelial Cells (ECs) has shown promise for vascular augmentation, but it face challenges in hostile tissue microenvironments, including poor cell survival and limited efficacy. In this study, the mitochondria of human BMSCs are isolated and transplanted to BMSCs from the same batch and passage number (BMSCsmito). The transplanted mitochondria significantly boosted the ability of BMSCsmito-ECs to promote angiogenesis, as assessed by in vitro tube formation and spheroid sprouting assays, as well as in vivo transplantation experiments in balb/c mouse and SD rat models. The Dll4-Notch1 signaling pathway is found to play a key role in BMSCsmito-induced endothelial tube formation. Co-transplanting BMSCsmito with ECs in a rat cranial bone defect significantly improves functional vascular network formation, and improve bone repair outcomes. These findings thus highlight that mitochondrial transplantation, by acting through the DLL4-Notch1 signaling pathway, represents a promising therapeutic strategy for enhancing angiogenesis and improving bone repair. Hence, mitochondrial transplantation to BMSCS as a therapeutic approach for promoting angiogenesis offers valuable insights and holds much promise for innovative regenerative medicine therapies.

4.
Acta Biomater ; 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39089349

ABSTRACT

The cell (plasma) membrane is enriched with numerous receptors, ligands, enzymes, and phospholipids that play important roles in cell-cell and cell-extracellular matrix interactions governing, for instance, tissue development and repair. We previously showed that plasma membrane nanofragments (PMNFs) act as nucleation sites for bone formation in vivo, and induce in vitro mineralization within 1 day. In this study, we optimized the methods for generating, isolating, and applying PMNFs as a cell-free therapeutic to expedite bone defect repair. The PMNFs were isolated from different mouse cell lines (chondrocytes, osteoblasts, and fibroblasts), pre-conditioned, lyophilized, and subsequently transplanted into 2 mm critical-sized calvarial defects in mice (n = 75). The PMNFs from chondrocytes, following a 3-day pre-incubation, significantly accelerated bone repair within 2 weeks, through a coordinated attraction of macrophages, endothelial cells, and osteoblasts to the healing site. In vitro experiments confirmed that PMNFs enhanced cell adhesion. Comparison of the PMNF efficacy with phosphatidylserine, amorphous calcium phosphate (ACP), and living cells confirmed the unique ability of PMNFs to promote accelerated bone repair. Importantly, PMNFs promoted nearly complete integration of the regenerated bone with native tissue after 6 weeks (% non-integrated bone area = 15.02), contrasting with the partial integration (% non-integrated bone area = 56.10; p < 0.01, Student's test) with transplantation of ACP. Vickers microhardness tests demonstrated that the regenerated bone after 6 weeks (30.10 ± 1.75) exhibited hardness similar to native bone (31.07 ± 2.46). In conclusion, this is the first study to demonstrate that cell membrane can be a promising cell-free material with multifaceted biofunctional properties that promote accelerated bone repair. STATEMENT OF SIGNIFICANCE.

5.
J Biomater Appl ; : 8853282241268676, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39151162

ABSTRACT

One of the critical factors that determines the biological properties of scaffolds is their structure. Due to the mechanical and structural discrepancies between the target bone and implants, the poor internal architecture design and difficulty in degradation of conventional bone implants may cause several adverse outcomes. To date, many scaffolds, such as 3-D printed sandwich structures, have been successfully developed for the repair of bone defects; however, the steps of these methods are complex and costly. Hydrogels have emerged as a unique scaffold material for repairing bone defects because of their good biocompatibility and excellent physicochemical properties. However, studies exploring bioinspired hydrogel scaffolds with hierarchical structures are scarce. More efforts are needed to incorporate bioinspired structures into hydrogel scaffolds to achieve optimal osteogenic properties. In this study, we developed a low-cost and easily available hydrogel matrix that mimicked the natural structure of the bone's porous sandwich to promote new bone growth and tissue integration. A comprehensive evaluation was conducted on the microstructure, swelling rate, and mechanical properties of this hydrogel. Furthermore, a 3D finite element analysis was employed to model the structure-property relationship. The results indicate that the sandwich-structured hydrogel is a promising scaffold material for bone injury repair, exhibiting enhanced compressive stress, elastic modulus, energy storage modulus, and superior force transmission.

6.
J Biomater Appl ; : 8853282241274528, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39152927

ABSTRACT

Piezoelectric ceramics are piezoelectric materials with polycrystalline structure and have been widely used in many fields such as medical imaging and sound sensors. As knowledge about this kind of material develops, researchers find piezoelectric ceramics possess favorable piezoelectricity, biocompatibility, mechanical properties, porous structure and antibacterial effect and endeavor to apply piezoelectric ceramics to the field of bone tissue engineering. However, clinically no piezoelectric ceramics have been exercised so far. Therefore, in this paper we present a comprehensive review of the research and development of various piezoelectric ceramics including barium titanate, potassium sodium niobate and zinc oxide ceramics and aims to explore the application of piezoelectric ceramics in bone regeneration by providing a detailed overview of the current knowledge and research of piezoelectric ceramics in bone tissue regeneration.

7.
J Mech Behav Biomed Mater ; 158: 106674, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39088942

ABSTRACT

Alveolar bone defects caused by tumor, trauma and inflammation can lead to the loss of oral function and complicate denture restoration. Currently, guided bone regeneration (GBR) barrier membranes for repairing bone defect cannot effectively promote bone regeneration due to their unstable degradation rate and poor antibacterial properties. Furthermore, they require additional tailoring before implantation. Therefore, this study developed a visible light-curing hydrogel membrane (CF-Cu) comprising methacrylated carboxymethyl chitosan (CMCS-MA), silk fibroin (SF), and copper nanoparticles (Cu NPs) to address these shortcomings of commercial membranes. The CF-Cu hydrogel, characterized by scanning electron microscopy (SEM), a universal testing machine, and swelling and degradation tests, demonstrated a smooth porous network structure, suitable swelling ratio, biodegradability, and enhanced mechanical strength. Cytotoxicity and hemolysis tests in vitro demonstrated excellent cyto- and hemo-compatibility of the CF-Cu hydrogel extracts. Additionally, evaluation of antibacterial properties in vitro, including colony forming unit (CFU) counts, MTT assays, and live/dead fluorescence staining, showed that the CF-Cu hydrogel exhibited excellent antibacterial properties, inhibiting over 80% of S. aureus, S. mutans, and P. gingivalis with CF-1Cu hydrogel compared to the control group. Moreover, evaluation of osteogenic differentiation of rBMSCs in vitro suggested that the CF-1Cu hydrogel significantly improved alkaline phosphatase (ALP) activity and the mineralization of extracellular matrix, up-regulating the expressions of osteogenesis-related genes (Runx2, ALP, Col-1, OPN and BSP). In summary, these results indicated that CF-1Cu hydrogel exhibited excellent cytocompatibility, antibacterial and osteogenic properties in vitro. Therefore, the CF-1Cu hydrogel holds potential as a viable material for application in GBR procedures aimed at addressing bone defects.

8.
Theranostics ; 14(11): 4438-4461, 2024.
Article in English | MEDLINE | ID: mdl-39113795

ABSTRACT

The high incidence of bone defect-related diseases caused by trauma, infection, and tumor resection has greatly stimulated research in the field of bone regeneration. Generally, bone healing is a long and complicated process wherein manipulating the biological activity of interventional scaffolds to support long-term bone regeneration is significant for treating bone-related diseases. It has been reported that some physical cues can act as growth factor substitutes to promote osteogenesis through continuous activation of endogenous signaling pathways. This review focuses on the latest progress in bone repair by remote actuation and on-demand activation of biomaterials pre-incorporated with physical cues (heat, electricity, and magnetism). As an alternative method to treat bone defects, physical cues show many advantages, including effectiveness, noninvasiveness, and remote manipulation. First, we introduce the impact of different physical cues on bone repair and potential internal regulatory mechanisms. Subsequently, biomaterials that mediate various physical cues in bone repair and their respective characteristics are summarized. Additionally, challenges are discussed, aiming to provide new insights and suggestions for developing intelligent biomaterials to treat bone defects and promote clinical translation.


Subject(s)
Biocompatible Materials , Bone Regeneration , Tissue Scaffolds , Bone Regeneration/drug effects , Biocompatible Materials/chemistry , Humans , Animals , Tissue Scaffolds/chemistry , Osteogenesis/drug effects , Tissue Engineering/methods , Bone and Bones/metabolism
9.
J Biophotonics ; : e202400066, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39048930

ABSTRACT

The aim of this study was to verify the effectiveness of attenuated total reflectance-fourier transform infrared (ATR-FTIR) spectroscopy in the characterization of bone repair in mandibular osteotomy using erbium, chromium-doped yttrium, scandium, gallium and garnet (Er,Cr:YSGG) laser and multilaminate drill on each side. Two mandible bone fragments were removed from 30 rabbits, and the process of bone repair was studied immediately, 3, 7, 15, 21, and 28 days after the surgery. The histological analysis allowed detecting differences in the early stages of tissue repair after bone cutting performed with the Er,Cr:YSGG laser or multilaminate drill. The ATR-FTIR spectroscopy technique was sensitive to changes in the organic content of bone tissue repair process.

10.
J Biomater Appl ; : 8853282241268683, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39056481

ABSTRACT

The design and construction of a new and excellent synthetic graft is of great significance in the field of bone defect repair and reconstruction. In this study, a dopamine modified chitosan hydrogel doped with Cu ions with a mild photothermal effect was designed to provide a better microenvironment to advance the bone repair via promote the angiogenesis and osteogenesis. Characterizations showed the successful synthesis of the material while it also presented excellent biocompatibility and mild photothermal effect under the irradiation of near-infrared light. Further, it could enhance the angiogenesis of HUVECs cells through promoting the ability of migration and tube formation and enhance the osteogenic differentiation of MC3T3-E1 cells via increasing the content of vital osteogenic factors including Runx2, Col-1, OPN, OCN, OSX, etc. The in vivo experiment also testified that it could promote the bone defect repair in rat models. These results indicate the multifunctional hydrogel is an ideal material for the treatment of bone defects and has good clinical application potential.

11.
Tissue Cell ; 89: 102422, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39003912

ABSTRACT

Tumour necrosis factor alpha (TNF-α) is a pleiotropic cytokine synthesised primarily by mononuclear cells; it has a potent pro-inflammatory effect, playing a crucial role in metabolic, immune, and inflammatory diseases. This cytokine has been studied in various biological systems. In bone tissue, TNF-α plays an integral role in skeletal disorders such as osteoporosis, fracture repair and rheumatoid arthritis through its involvement in regulating the balance between osteoblasts and osteoclasts, mediating inflammatory responses, promoting angiogenesis and exacerbating synovial proliferation. The biological effect TNF-α exerts in this context is determined by a combination of the signalling pathway it activates, the type of receptor it binds, and the concentration and duration of exposure. This review summarises the participation and pathophysiological role of TNF-α in osteoporosis, bone damage repair, chronic immunoinflammatory bone disease and spinal cord injury, and discusses its main mechanisms.


Subject(s)
Osteoporosis , Tumor Necrosis Factor-alpha , Humans , Osteoporosis/metabolism , Osteoporosis/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Inflammation/metabolism , Inflammation/pathology , Osteoblasts/metabolism , Bone Diseases/metabolism , Bone Diseases/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Signal Transduction
12.
Biotechnol J ; 19(7): e2300751, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987220

ABSTRACT

The compatibility of bone graft substitutes (BGS) with mesenchymal stem cells (MSCs) is an important parameter to consider for their use in repairing bone defects as it eventually affects the clinical outcome. In the present study, a few commercially available BGS - ß-tricalcium phosphate (ß-TCP), calcium sulfate, gelatin sponge, and different forms of hydroxyapatite (HAP) were screened for their interactions with MSCs from adipose tissue (ADSCs). It was demonstrated that HAP block favorably supported ADSC viability, morphology, migration, and differentiation compared to other scaffolds. The results strongly suggest the importance of preclinical evaluation of bone scaffolds for their cellular compatibility. Furthermore, the bone regenerative potential of HAP block with ADSCs was evaluated in an ex vivo bone defect model developed using patient derived trabecular bone explants. The explants were cultured for 45 days in vitro and bone formation was assessed by expression of osteogenic genes, ALP secretion, and high resolution computed tomography. Our findings confirmed active bone repair process in ex vivo settings. Addition of ADSCs significantly accelerated the repair process and improved bone microarchitecture. This ex vivo bone defect model can emerge as a viable alternative to animal experimentation and also as a potent tool to evaluate patient specific bone therapeutics under controlled conditions.


Subject(s)
Adipose Tissue , Bone Regeneration , Cell Differentiation , Mesenchymal Stem Cells , Tissue Engineering , Tissue Scaffolds , Humans , Adipose Tissue/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Mesenchymal Stem Cells/cytology , Femur Head , Osteogenesis , Cells, Cultured , Bone Substitutes/chemistry , Durapatite/chemistry , Calcium Phosphates/chemistry
13.
Adv Healthc Mater ; : e2401275, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38979868

ABSTRACT

Compromised osteogenesis and angiogenesis is the character of stem cell senescence, which brought difficulties for bone defects repairing in senescent microenvironment. As the most abundant bone-related miRNA, miRNA-21-5p plays a crucial role in inducing osteogenic and angiogenic differentiation. However, highly efficient miR-21-5p delivery still confronts challenges including poor cellular uptake and easy degradation. Herein, TDN-miR-21-5p nanocomplex is constructed based on DNA tetrahedral (TDN) and has great potential in promoting osteogenesis and alleviating senescence of senescent bone marrow stem cells (O-BMSCs), simultaneously enhancing angiogenic capacity of senescent endothelial progenitor cells (O-EPCs). Of note, the activation of AKT and Erk signaling pathway may direct regulatory mechanism of TDN-miR-21-5p mediated osteogenesis and senescence of O-BMSCs. Also, TDN-miR-21-5p can indirectly mediate osteogenesis and senescence of O-BMSCs through pro-angiogenic growth factors secreted from O-EPCs. In addition, gelatin methacryloyl (GelMA) hydrogels are mixed with TDN and TDN-miR-21-5p to fabricate delivery scaffolds. TDN-miR-21-5p@GelMA scaffold exhibits greater bone repair with increased expression of osteogenic- and angiogenic-related markers in senescent critical-size cranial defects in vivo. Collectively, TDN-miR-21-5p can alleviate senescence and induce osteogenesis and angiogenesis in senescent microenvironment, which provides a novel candidate strategy for senescent bone repair and widen clinical application of TDNs-based gene therapy.

14.
Article in English | MEDLINE | ID: mdl-39033545

ABSTRACT

In situ 3D printing is attractive for the direct repair of bone defects in underdeveloped countries and in emergency situations. So far, the lack of an interesting method to produce filament using FDA-approved biopolymers and nanoceramics combined with a portable strategy limits the use of in situ 3D printing. Herein, we investigated the osseointegration of new nanocomposite filaments based on polylactic acid (PLA), laponite (Lap), and hydroxyapatite (Hap) printed directly at the site of the bone defect in rats using a portable 3D printer. The filaments were produced using a single-screw extruder (L/D = 26), without the addition of solvents that can promote the toxicity of the materials. In vitro performance was evaluated in the cell differentiation process with mesenchymal stem cells (MSC) by an alkaline phosphatase activity test and visualization of mineralization nodules; a cell viability test and total protein dosage were performed to evaluate cytotoxicity. For the in vivo analysis, the PLA/Lap composite filaments with a diameter of 1.75 mm were printed directly into bone defects of Wistar rats using a commercially available portable 3D printer. Based on the in vitro and in vivo results, the in situ 3D printing technique followed by rapid cooling proved to be promising for bone tissue engineering. The absence of fibrous encapsulation and inflammatory processes became a good indicator of effectiveness in terms of biocompatibility parameters and bone tissue formation, and the use of the portable 3D printer showed a significant advantage in the application of this material by in situ printing.

15.
ACS Appl Mater Interfaces ; 16(28): 35964-35984, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38968558

ABSTRACT

Developing a neurovascular bone repair scaffold with an appropriate mechanical strength remains a challenge. Calcium phosphate (CaP) is similar to human bone, but its scaffolds are inherently brittle and inactive, which require recombination with active ions and polymers for bioactivity and suitable strength. This work discussed the synthesis of amorphous magnesium-calcium pyrophosphate (AMCP) and the subsequent development of a humidity-responsive AMCP/cassava starch (CS) scaffold. The scaffold demonstrated enhanced mechanical properties by strengthening the intermolecular hydrogen bonds and ionic bonds between AMCP and CS during the gelatinization and freeze-thawing processes. The release of active ions was rapid initially and stabilized into a long-term stable release after 3 days, which is well-matched with new bone growth. The release of pyrophosphate ions endowed the scaffold with antibacterial properties. At the cellular level, the released active ions simultaneously promoted the proliferation and mineralization of osteoblasts, the proliferation and migration of endothelial cells, and the proliferation of Schwann cells. At the animal level, the scaffold was demonstrated to promote vascular growth and peripheral nerve regeneration in a rat skull defect experiment, ultimately resulting in the significant and rapid repair of bone defects. The construction of the AMCP/CS scaffold offers practical suggestions and references for neurovascular bone repair.


Subject(s)
Bone Regeneration , Starch , Tissue Scaffolds , Animals , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Rats , Starch/chemistry , Humidity , Humans , Cell Proliferation/drug effects , Rats, Sprague-Dawley , Diphosphates/chemistry , Diphosphates/pharmacology , Osteoblasts/drug effects , Osteoblasts/cytology , Calcium Pyrophosphate/chemistry , Calcium Pyrophosphate/pharmacology , Schwann Cells/drug effects , Schwann Cells/cytology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Skull/drug effects
16.
Int J Pharm ; 661: 124414, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38960344

ABSTRACT

Bulleyaconitine A (BLA) is a promising candidate for treating rheumatoid arthritis (RA) with diverse pharmacological activities, including anti-inflammatory, analgesic and bone repair. Herein, the long-acting bulleyaconitine A microspheres (BLA-MS) were developed to treat RA comprehensively by forming drug reservoirs in joint cavities. The BLA-MS were prepared by emulsion/solvent evaporation method. The particle size and distribution were assessed by SEM. The crystalline state was investigated by DSC and PXRD. The drug loading (DL), encapsulation efficiency (EE) and cumulative release in vitro were determined by HPLC. The DL and EE were 23.93 ± 0.38 % and 95.73 ± 1.56 % respectively, and the cumulative release was up to 69 days with a stable release curve. The pharmacodynamic results in collagen induced arthritis (CIA) rats showed a noticeable reduction in paw thickness (5.66 ± 0.32 mm), and the decreasing expression level of PGE2, TNF-α and IL-6 which diminished the infiltration of inflammatory cells, thereby alleviating the progression of erosion and repairing the damaged bones (BV/TV (Bone Volume / Total Volume): 81.97 %, BS/BV (Bone Surface / Bone Volume): 6.08 mm-1). In conclusion, intra-articular injection of BLA-MS should have a promising application in the treatment of RA and may achieve clinical transformation in the future.


Subject(s)
Aconitine , Arthritis, Experimental , Arthritis, Rheumatoid , Drug Liberation , Microspheres , Animals , Aconitine/analogs & derivatives , Aconitine/administration & dosage , Aconitine/chemistry , Aconitine/pharmacokinetics , Injections, Intra-Articular , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Male , Rats , Particle Size , Delayed-Action Preparations , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/chemistry
17.
Stem Cells Transl Med ; 13(8): 791-802, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-38986535

ABSTRACT

Platelet-derived growth factor receptor α (PDGFRα) is often considered as a general marker of mesenchymal cells and fibroblasts, but also shows expression in a portion of osteoprogenitor cells. Within the skeleton, Pdgfrα+ mesenchymal cells have been identified in bone marrow and periosteum of long bones, where they play a crucial role in participating in fracture repair. A similar examination of Pdgfrα+ cells in calvarial bone healing has not been examined. Here, we utilize Pdgfrα-CreERTM;mT/mG reporter animals to examine the contribution of Pdgfrα+ mesenchymal cells to calvarial bone repair through histology and single-cell RNA sequencing (scRNA-Seq). Results showed that Pdgfrα+ mesenchymal cells are present in several cell clusters by scRNA-Seq, and by histology a dramatic increase in Pdgfrα+ cells populated the defect site at early timepoints to give rise to healed bone tissue overtime. Notably, diphtheria toxin-mediated ablation of Pdgfrα reporter+ cells resulted in significantly impaired calvarial bone healing. Our findings suggest that Pdgfrα-expressing cells within the calvarial niche play a critical role in the process of calvarial bone repair.


Subject(s)
Receptor, Platelet-Derived Growth Factor alpha , Skull , Animals , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Mice , Skull/metabolism , Skull/injuries , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Bone Regeneration/physiology
18.
Regen Biomater ; 11: rbae078, 2024.
Article in English | MEDLINE | ID: mdl-39055303

ABSTRACT

The intricate nature of oral-maxillofacial structure and function, coupled with the dynamic oral bacterial environment, presents formidable obstacles in addressing the repair and regeneration of oral-maxillofacial bone defects. Numerous characteristics should be noticed in oral-maxillofacial bone repair, such as irregular morphology of bone defects, homeostasis between hosts and microorganisms in the oral cavity and complex periodontal structures that facilitate epithelial ingrowth. Therefore, oral-maxillofacial bone repair necessitates restoration materials that adhere to stringent and specific demands. This review starts with exploring these particular requirements by introducing the particular characteristics of oral-maxillofacial bones and then summarizes the classifications of current bone repair materials in respect of composition and structure. Additionally, we discuss the modifications in current bone repair materials including improving mechanical properties, optimizing surface topography and pore structure and adding bioactive components such as elements, compounds, cells and their derivatives. Ultimately, we organize a range of potential optimization strategies and future perspectives for enhancing oral-maxillofacial bone repair materials, including physical environment manipulation, oral microbial homeostasis modulation, osteo-immune regulation, smart stimuli-responsive strategies and multifaceted approach for poly-pathic treatment, in the hope of providing some insights for researchers in this field. In summary, this review analyzes the complex demands of oral-maxillofacial bone repair, especially for periodontal and alveolar bone, concludes multifaceted strategies for corresponding biomaterials and aims to inspire future research in the pursuit of more effective treatment outcomes.

19.
J Orthop Translat ; 47: 132-143, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39027342

ABSTRACT

Background: A pivotal determinant for the success of tissue regeneration lies in the establishment of sufficient vasculature. Utilizing autologous tissue grafts from donors offers the dual advantage of mitigating the risk of disease transmission and circumventing the necessity for post-transplant immunosuppression, rendering it an exemplary vascularization strategy. Among the various potential autologous donors, adipose tissue emerges as a particularly auspicious source, being both widely available and compositionally rich. Notably, adipose-derived microvascular fragments (ad-MVFs) are a promising candidate for vascularization. ad-MVFs can be isolated from adipose tissue in a short period of time and show high vascularized capacity. In this study, we extracted ad-MVFs from adipose tissue and utilized their strong angiogenic ability to accelerate bone repair by promoting vascularization. Methods: ad-MVFs were extracted from the rat epididymis using enzymatic hydrolysis. To preserve the integrity of the blood vessels, gelatin methacryloyl (GelMA) hydrogel was chosen as the carrier for ad-MVFs in three-dimensional (3D) culture. The ad-MVFs were cultured directly on the well plates for two-dimensional (2D) culture as a control. The morphology of ad-MVFs was observed under both 2D and 3D cultures, and the release levels of vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP-2) were assessed under both culture conditions. In vitro studies investigated the impact of ad-MVFs/GelMA hydrogel on the toxicity, osteoblastic activity, and mineralization of rat bone marrow mesenchymal stem cells (rBMSCs), along with the examination of osteogenic gene and protein expression. In vivo experiments involved implanting the ad-MVFs/GelMA hydrogel into critical-size skull defects in rats, and its osteogenic ability was evaluated through radiographic and histological methods. Results: ad-MVFs were successfully isolated from rat adipose tissue. When cultured under 2D conditions, ad-MVFs exhibited a gradual disintegration and loss of their original vascular morphology. Compared with 2D culture, ad-MVFs can not only maintain the original vascular morphology, but also connect into a network in hydrogel under 3D culture condition. Moreover, the release levels of VEGF and BMP-2 were significantly higher than those in 2D culture. Moreover, the ad-MVFs/GelMA hydrogel exhibited superior osteoinductive activity. After implanting into the skull defect of rats, the ad-MVFs/GelMA hydrogel showed obvious effects for angiogenesis and osteogenesis. The translational potential of this article: The utilization of autologous adipose tissue as a donor presents a more direct route toward clinical translation. Anticipated future clinical applications envision the transformation of discarded adipose tissue into a valuable resource for personalized tissue repair, thereby realizing a paradigm shift in the utilization of this abundant biological material.

20.
Regen Biomater ; 11: rbae070, 2024.
Article in English | MEDLINE | ID: mdl-39022124

ABSTRACT

A hybrid material possessing both componential and structural imitation of bone tissue is the preferable composites for bone defect repair. Inspired by the microarchitecture of native bone, this work synthesized in vitro a functional mineralized collagen fibril (MCF) material by utilizing the method of in situ co-precipitation, which was designed to proceed in the presence of Astragalus polysaccharide (APS), thus achieving APS load within the biomineralized collagen-Astragalus polysaccharide (MCAPS) fibrils. Transmission electron microscope (TEM), selected area electron diffraction (SAED) and scanning electronic microscopy (SEM) identified the details of the intrafibrillar mineralization of the MCAPS fibrils, almost mimicking the secondary level of bone tissue microstructure. A relatively uniform and continuous mineral layer formed on and within all collagen fibrils and the mineral phase was identified as typical weak-crystalline hydroxyapatite (HA) with a Ca/P ratio of about 1.53. The proliferation of bone marrow-derived mesenchymal stem cells (BMSC) and mouse embryo osteoblast precursor cells (MC3T3-E1) obtained a significant promotion by MCAPS. As for the osteogenic properties of MCAPS, a distinct increase in the alkaline phosphatase (ALP) activity and the number of calcium nodules (CN) in BMSC and MC3T3-E1 was detected. The up-regulation of three osteogenic-related genes of RUNX-2, BMP-2 and OCN were confirmed via reverse transcription-quantitative polymerase chain reaction (RT-qPCR) to further verify the osteogenic performance promotion of MCAPS. A period of 14 days of culture demonstrated that MCAPS-L exhibited a preferable efficacy in enhancing ALP activity and CN quantity, as well as in promoting the expression of osteogenic-related genes over MCAPS-M and MCAPS-H, indicating that a lower dose of APS within the material of MCAPS is more appropriate for its osteogenesis promotion properties.

SELECTION OF CITATIONS
SEARCH DETAIL