Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.110
Filter
1.
J Nanobiotechnology ; 22(1): 577, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300539

ABSTRACT

Treating bone defect concomitant with microbial infection poses a formidable clinical challenge. Addressing this dilemma necessitates the implementation of biomaterials exhibiting dual capabilities in anti-bacteria and bone regeneration. Of particular significance is the altered microenvironment observed in infected bones, characterized by acidity, inflammation, and an abundance of reactive oxygen species (ROS). These conditions, while challenging, present an opportunity for therapeutic intervention in the context of contaminated bone defects. In this study, we developed an oriented composite scaffold containing copper-coated manganese dioxide (MnO2) nanoparticles loaded with parathyroid hormone (PMPC/Gelatin). The characteristics of these scaffolds were meticulously evaluated and confirmed the high sensitivity to H+, responsive drug release and ROS elimination. In vitro antibacterial analysis underscored the remarkable ability of PMPC/Gelatin scaffolds to substantially suppressed bacterial proliferation and colony formation. Furthermore, this nontoxic material demonstrated efficacy in mitigating ROS levels, thereby fostering osteogenic differentiation of bone marrow mesenchymal stem cells and enhancing angiogenic ability. Subsequently, the infected models of bone defects in rat skulls were established to investigate the effects of composite scaffolds on anti-bacteria and bone formation in vivo. The PMPC/Gelatin treatment exhibited excellent antibacterial activity, coupled with enhanced vascularization and osteogenesis at the defect sites. These compelling findings affirm that the PMPC/Gelatin composite scaffold represents a promising avenue for anti-bacteria and bone regeneration.


Subject(s)
Anti-Bacterial Agents , Bone Regeneration , Gelatin , Manganese Compounds , Mesenchymal Stem Cells , Osteogenesis , Oxides , Rats, Sprague-Dawley , Reactive Oxygen Species , Tissue Scaffolds , Animals , Osteogenesis/drug effects , Tissue Scaffolds/chemistry , Rats , Bone Regeneration/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Mesenchymal Stem Cells/drug effects , Reactive Oxygen Species/metabolism , Gelatin/chemistry , Oxides/chemistry , Oxides/pharmacology , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Male , Skull/drug effects , Nanoparticles/chemistry , Copper/chemistry , Copper/pharmacology , Cell Differentiation/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
2.
Sci Rep ; 14(1): 20848, 2024 09 06.
Article in English | MEDLINE | ID: mdl-39242756

ABSTRACT

The development of synthetic bone substitutes that equal or exceed the efficacy of autologous graft remains challenging. In this study, a rat calvarial defect model was used as a reference to investigate the influence of composition and architecture of 3D-printed cement, with or without bioactives, on tissue regeneration. Printable cement pastes were formulated by combining hyaluronic acid and cement precursors. Cementitious scaffolds were printed with 3 different patterns. After 7 weeks of implantation with or without bone marrow, multiparametric qualitative and quantitative assessments were performed using µCT, SEM, and histology. None of the set-up strategies was as efficient as autologous cancellous bone graft to repair calvarial defects. Nonetheless, the presence of scaffold improved the skull vault closure, particularly when the scaffold was soaked in total bone marrow before implantation. No significant effect of scaffold macro-architecture was observed on tissue mineralization. Magnesium phosphate-based scaffolds (MgP) seemed to induce higher bone formation than their calcium-phosphate-based counterparts. They also displayed a quicker biodegradation and sparse remaining material was found after 7 weeks of implantation. Although further improvements are required to reach clinical settings, this study demonstrated the potential of organo-mineral cements for bone regeneration and highlighted the peculiar properties of MgP-based cements.


Subject(s)
Bone Regeneration , Printing, Three-Dimensional , Skull , Tissue Scaffolds , Animals , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Rats , Skull/drug effects , Bone Substitutes/chemistry , Bone Substitutes/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Male , Bone Cements/pharmacology , Bone Cements/chemistry , Phosphates/chemistry , Osteogenesis/drug effects , X-Ray Microtomography , Magnesium Compounds
3.
J Nanobiotechnology ; 22(1): 539, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237993

ABSTRACT

Maxillofacial bone defects can severely impact quality of life by impairing physiological functions such as chewing, breathing, swallowing, and pronunciation. Polyether ether ketone (PEEK) is commonly used for the repair of maxillofacial defects due to its mechanical adaptability, while its osteogenic properties still need refinement. Herein, we have utilized the piezoelectric effect exhibited by barium titanate (BTO) under low-intensity pulsed ultrasound (LIPUS) to develop an ultrasound responsive PEEK (PDA@BTO-SPEEK, PBSP) through the mediating effect of polydopamine (PDA), for repairing maxillofacial bone defects. After modification by PDA@BTO, PBSP possesses better hydrophilicity, which is conducive to cell growth and adhesion. Simultaneously, by virtue of the piezoelectric characteristics of BTO, PBSP obtains a piezoelectric coefficient that matches the bone cortex. Notably, when PBSP is stimulated by LIPUS, it can generate stable electricity and effectively accelerate the osteogenic differentiation of osteoblasts through the regulation of the Piezo1-induced calcium (Ca2+) influx and Akt/GSK3ß/ß-catenin pathway. In addition, PBSP presents satisfactory therapeutic effects in rat skull defect models, and its osteogenic efficiency can be further improved under LIPUS stimulation with high tissue penetration. Collectively, PBSP + LIPUS exhibits great potential as a promising alternative strategy for the repair of maxillofacial bone defects.


Subject(s)
Benzophenones , Glycogen Synthase Kinase 3 beta , Ketones , Osteogenesis , Polyethylene Glycols , Polymers , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , beta Catenin , Animals , Glycogen Synthase Kinase 3 beta/metabolism , Polymers/chemistry , Osteogenesis/drug effects , Rats , Polyethylene Glycols/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Ketones/chemistry , Ketones/pharmacology , beta Catenin/metabolism , Cell Differentiation/drug effects , Osteoblasts/drug effects , Ultrasonic Waves , Indoles/chemistry , Indoles/pharmacology , Male , Signal Transduction/drug effects , Skull/drug effects , Titanium/chemistry , Titanium/pharmacology , Bone Regeneration/drug effects
4.
Biomed Mater ; 19(5)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39094613

ABSTRACT

The design of three-dimensional (3D) scaffolds should focus on creating highly porous, 3D structures with an interconnected pore network that supports cell growth. The scaffold's pore interconnectivity is directly linked to vascularization, cell seeding, guided cell migration, and transportation of nutrients and metabolic waste. In this study, different types of food flavors including monosodium glutamate, sugar, and sodium chloride were used as the porogens along with PCL/PVP blend polymer for solvent casting/particulate leaching method. The morphology, porosity, interconnectivity, chemical composition, water absorption, and mechanical properties of the fabricated scaffolds are carefully characterized. The scaffolds are biocompatible in bothin vitroandin vivoexperiments and do not trigger any inflammatory response while enhancing new bone formation and vascularization in rabbit calvaria critical-sized defects. The new bone merges and becomes denser along with the experiment timeline. The results indicate that the 3D PCL/PVP scaffolds, using monosodium glutamate as porogen, exhibited suitable biological performance and held promise for bone tissue engineering in oral and maxillofacial surgery.


Subject(s)
Biocompatible Materials , Sodium Glutamate , Solvents , Tissue Engineering , Tissue Scaffolds , Animals , Tissue Scaffolds/chemistry , Rabbits , Tissue Engineering/methods , Porosity , Solvents/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Polyesters/chemistry , Materials Testing , Skull/drug effects , Polyvinyls/chemistry , Bone Regeneration/drug effects , Osteogenesis/drug effects , Bone Substitutes/chemistry , Bone Substitutes/pharmacology , Bone and Bones/metabolism
5.
Int J Nanomedicine ; 19: 8309-8336, 2024.
Article in English | MEDLINE | ID: mdl-39161358

ABSTRACT

Purpose: The treatment of craniofacial bone defects caused by trauma, tumors, and infectious and degenerative diseases is a significant issue in current clinical practice. Following the rapid development of bone tissue engineering (BTE) in the last decade, bioactive scaffolds coupled with multifunctional properties are in high demand with regard to effective therapy for bone defects. Herein, an innovative bone scaffold consisting of GO/Cu nanoderivatives and GelMA-based organic-inorganic hybrids was reported for repairing full-thickness calvarial bone defect. Methods: In this study, motivated by the versatile biological functions of nanomaterials and synthetic hydrogels, copper nanoparticle (CuNP)-decorated graphene oxide (GO) nanosheets (GO/Cu) were combined with methacrylated gelatin (GelMA)-based organic-inorganic hybrids to construct porous bone scaffolds that mimic the extracellular matrix (ECM) of bone tissues by photocrosslinking. The material characterizations, in vitro cytocompatibility, macrophage polarization and osteogenesis of the biohybrid hydrogel scaffolds were investigated, and two different animal models (BALB/c mice and SD rats) were established to further confirm the in vivo neovascularization, macrophage recruitment, biocompatibility, biosafety and bone regenerative potential. Results: We found that GO/Cu-functionalized GelMA/ß-TCP hydrogel scaffolds exhibited evidently promoted osteogenic activities, M2 type macrophage polarization, increased secretion of anti-inflammatory factors and excellent cytocompatibility, with favorable surface characteristics and sustainable release of Cu2+. Additionally, improved neovascularization, macrophage recruitment and tissue integration were found in mice implanted with the bioactive hydrogels. More importantly, the observations of microCT reconstruction and histological analysis in a calvarial bone defect model in rats treated with GO/Cu-incorporated hydrogel scaffolds demonstrated significantly increased bone morphometric values and newly formed bone tissues, indicating accelerated bone healing. Conclusion: Taken together, this BTE-based bone repair strategy provides a promising and feasible method for constructing multifunctional GO/Cu nanocomposite-incorporated biohybrid hydrogel scaffolds with facilitated osteogenesis, angiogenesis and immunoregulation in one system, with the optimization of material properties and biosafety, it thereby demonstrates great application potential for correcting craniofacial bone defects in future clinical scenarios.


Subject(s)
Bone Regeneration , Copper , Graphite , Hydrogels , Rats, Sprague-Dawley , Skull , Tissue Engineering , Tissue Scaffolds , Animals , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Copper/chemistry , Copper/pharmacology , Graphite/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Skull/drug effects , Skull/injuries , Rats , Mice , Tissue Engineering/methods , Osteogenesis/drug effects , Mice, Inbred BALB C , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Male , Metal Nanoparticles/chemistry , Nanostructures/chemistry , Gelatin/chemistry , RAW 264.7 Cells
6.
Braz J Med Biol Res ; 57: e13913, 2024.
Article in English | MEDLINE | ID: mdl-39166608

ABSTRACT

Considering the lack of consensus related to the impact of selective IL-6 receptor inhibition on bone remodeling and the scarcity of reports, especially on large bone defects, this study proposed to evaluate the biological impact of the selective inhibitor of interleukin-6 receptor (tocilizumab) in an experimental model of critical calvarial defect in rats. In this preclinical and in vivo study, 24 male Wistar rats were randomly divided into two groups (n=12/group): defect treated with collagen sponge (CG) and defect treated with collagen sponge associated with 2 mg/kg tocilizumab (TCZ). The defect in the parietal bone was created using an 8-mm diameter trephine drill. After 90 days, the animals were euthanized, and tissue samples (skull caps) were evaluated through micro-CT, histological, immunohistochemistry, cytokines, and RT-qPCR analyses. Tocilizumab reduced mononuclear inflammatory infiltration (P<0.05) and tumor necrosis factor (TNF)-α levels (P<0.01) and down-regulated tissue gene expression of BMP-2 (P<0.001), RUNX-2 (P<0.05), and interleukin (IL)-6 (P<0.05). Moreover, it promoted a stronger immunostaining of cathepsin and RANKL (P<0.05). Micro-CT and histological analyses revealed no impact on general bone formation (P>0.05). The bone cells (osteoblasts, osteoclasts, and osteocytes) in the defect area were similar in both groups (P>0.05). Tocilizumab reduced inflammatory cytokines, decreased osteogenic protein, and increased proteases in a critical bone defect in rats. Ninety days after the local application of tocilizumab in the cranial defect, we did not find a significant formation of bone tissue compared with a collagen sponge.


Subject(s)
Cytokines , Disease Models, Animal , Rats, Wistar , Receptors, Interleukin-6 , Skull , Animals , Male , Cytokines/metabolism , Receptors, Interleukin-6/antagonists & inhibitors , Skull/drug effects , Rats , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , X-Ray Microtomography , Peptide Hydrolases/metabolism , Immunohistochemistry , Random Allocation
7.
Commun Biol ; 7(1): 962, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122919

ABSTRACT

With increases in life expectancy, the number of patients requiring joint replacement therapy and experiencing periprosthetic osteolysis, the most common complication leading to implant failure, is growing or underestimated. In this study, we found that osteolysis progression and osteoclast differentiation in the surface of the skull bone of adult mice were accompanied by significant expansion of lymphatic vessels within bones. Using recombinant VEGF-C protein to activate VEGFR3 and promote proliferation of lymphatic vessels in bone, we counteracted excessive differentiation of osteoclasts and osteolysis caused by titanium alloy particles or inflammatory cytokines LPS/TNF-α. However, this effect was not observed in aged mice because adipogenically differentiated mesenchymal stem cells (MSCs) inhibited the response of lymphatic endothelial cells to agonist proteins. The addition of the JAK inhibitor ruxolitinib restored the response of lymphatic vessels to external stimuli in aged mice to protect against osteolysis progression. These findings suggest that inhibiting SASP secretion by adipogenically differentiated MSCs while activating lymphatic vessels in bone offers a new method to prevent periprosthetic osteolysis during joint replacement follow-up.


Subject(s)
Lymphatic Vessels , Mesenchymal Stem Cells , Osteolysis , Animals , Osteolysis/prevention & control , Mice , Lymphatic Vessels/drug effects , Lymphatic Vessels/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Aging , Mice, Inbred C57BL , Osteoclasts/metabolism , Osteoclasts/drug effects , Cell Differentiation/drug effects , Male , Phenotype , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics , Skull/pathology , Skull/drug effects , Vascular Endothelial Growth Factor Receptor-3/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Titanium
8.
J Nanobiotechnology ; 22(1): 525, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39217329

ABSTRACT

The complex anatomy and biology of craniofacial bones pose difficulties in their effective and precise reconstruction. Injectable hydrogels (IHs) with water-swollen networks are emerging as a shape-adaptive alternative for noninvasively rebuilding craniofacial bones. The advent of versatile nanomaterials (NMs) customizes IHs with strengthened mechanical properties and therapeutically favorable performance, presenting excellent contenders over traditional substitutes. Structurally, NM-reinforced IHs are energy dissipative and covalently crosslinked, providing the mechanics necessary to support craniofacial structures and physiological functions. Biofunctionally, incorporating unique NMs into IH expands a plethora of biological activities, including immunomodulatory, osteogenic, angiogenic, and antibacterial effects, further favoring controllable dynamic tissue regeneration. Mechanistically, NM-engineered IHs optimize the physical traits to direct cell responses, regulate intracellular signaling pathways, and control the release of biomolecules, collectively bestowing structure-induced features and multifunctionality. By encompassing state-of-the-art advances in NM-integrated IHs, this review offers a foundation for future clinical translation of craniofacial bone reconstruction.


Subject(s)
Bone Regeneration , Facial Bones , Hydrogels , Nanostructures , Tissue Engineering , Hydrogels/chemistry , Humans , Nanostructures/chemistry , Animals , Bone Regeneration/drug effects , Tissue Engineering/methods , Skull/drug effects , Osteogenesis/drug effects , Biocompatible Materials/chemistry , Tissue Scaffolds/chemistry
9.
Biomater Sci ; 12(16): 4226-4241, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38984522

ABSTRACT

Objectives: The technique of guided bone regeneration (GBR) has been widely used in the field of reconstructive dentistry to address hard tissue deficiency. The objective of this research was to manufacture a novel bi-layered asymmetric membrane that incorporates demineralized dentin matrix (DDM), a bioactive bone replacement derived from dentin, in order to achieve both soft tissue isolation and hard tissue regeneration simultaneously. Methods: DDM particles were harvested from healthy, caries-free permanent teeth. The electrospinning technique was utilized to synthesize bi-layered DDM-loaded PLGA/PLA (DPP) membranes. We analyzed the DPP bilayer membranes' surface topography, physicochemical properties and degradation ability. Rat skull critical size defects (CSDs) were constructed to investigate in vivo bone regeneration. Results: The synthesized DPP bilayer membranes possessed suitable surface characteristics, acceptable mechanical properties, good hydrophilicity, favorable apatite forming ability and suitable degradability. Micro-computed tomography (CT) showed significantly more new bone formation in the rat skull defects implanted with the DPP bilayer membranes. Histological evaluation further revealed that the bone was more mature with denser bone trabeculae. In addition, the DPP bilayer membrane significantly promoted the expression of the OCN matrix protein in vivo. Conclusions: The DPP bilayer membranes exhibited remarkable biological safety and osteogenic activity in vivo and showed potential as a prospective candidate for GBR applications in the future.


Subject(s)
Bone Regeneration , Dentin , Skull , Animals , Bone Regeneration/drug effects , Skull/injuries , Skull/pathology , Skull/diagnostic imaging , Skull/drug effects , Rats , Dentin/chemistry , Rats, Sprague-Dawley , Membranes, Artificial , Male , Wound Healing/drug effects , X-Ray Microtomography , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Tissue Scaffolds/chemistry , Osteogenesis/drug effects
10.
Tissue Eng Regen Med ; 21(7): 1093-1107, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38955905

ABSTRACT

BACKGROUND: This study aimed to identify glycine analogs conducive to the formation of cell-absorbable nanocomplexes, enhancing collagen synthesis and subsequent osteogenesis in combination with BMP2 for improved bone regeneration. METHODS: Glycine and its derivatives were assessed for their effects on osteogenic differentiation in MC3T3-E1 cells and human bone marrow mesenchymal stem cells (BMSCs) under osteogenic conditions or with BMP2. Osteogenic differentiation was assessed through alkaline phosphatase staining and real-time quantitative polymerase chain reaction (RT-qPCR). Nanocomplex formation was examined via scanning electron microscopy, circular dichroism, and ultraviolet-visible spectroscopy. In vivo osteogenic effects were validated using a mouse calvarial defect model, and bone regeneration was evaluated through micro-computed tomography and histomorphometric analysis. RESULTS: Glycine, glycine methyl ester, and glycinamide significantly enhanced collagen synthesis and ALP activity in conjunction with an osteogenic medium (OSM). GA emerged as the most effective inducer of osteoblast differentiation marker genes. Combining GA with BMP2 synergistically stimulated ALP activity and the expression of osteoblast markers in both cell lines. GA readily formed nanocomplexes, facilitating cellular uptake through strong electrostatic interactions. In an in vivo calvarial defect mouse model, the GA and BMP2 combination demonstrated enhanced bone volume, bone volume/tissue volume ratio, trabecular numbers, and mature bone formation compared to other combinations. CONCLUSION: GA and BMP2 synergistically promoted in vitro osteoblast differentiation and in vivo bone regeneration through nanocomplex formation. This combination holds therapeutic promise for individuals with bone defects, showcasing its potential for clinical intervention.


Subject(s)
Bone Morphogenetic Protein 2 , Bone Regeneration , Cell Differentiation , Osteoblasts , Osteogenesis , Skull , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Regeneration/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Cell Differentiation/drug effects , Mice , Skull/drug effects , Humans , Osteogenesis/drug effects , Glycine/pharmacology , Glycine/analogs & derivatives , Glycine/chemistry , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Disease Models, Animal , Cell Line
11.
PLoS One ; 19(7): e0307134, 2024.
Article in English | MEDLINE | ID: mdl-39024220

ABSTRACT

Selective serotonin re-uptake inhibitors (SSRI) widely used in the treatment of depression, anxiety, obsessive compulsive disorder, fibromyalgia, and migraine are among the most heavily prescribed drug class in the United States (US). Along with an overall rise in SSRI use, these medications are increasingly used by pregnant individuals and recent preclinical and clinical studies have indicated that SSRIs may increase the prevalence of congenital abnormalities and birth defects of the craniofacial region. Our group has developed pre-clinical models of study, including those that mimic the clinical use of SSRI in mice. Here we designed a study to interrogate a commonly prescribed SSRI drug, Citalopram, for its effects on craniofacial and dental development when introduced in utero. Pre-natal exposure to a clinically relevant dose of citalopram resulted in changes in craniofacial form identified by an increase in endocast volume in SSRI exposed postnatal day 15 mouse pups. More specifically, cranial length and synchondrosis length increased in SSRI exposed pups as compared to control pups of the same age. Additionally, growth center (synchondrosis) height and width and palate length and width decreased in SSRI exposed pups as compared to control un-exposed pups. Effects of SSRI on the molars was minimal. Craniofacial growth and development continue to be an area of interest in the investigation of in utero pharmaceutical drug exposure. Altogether these data indicate that prenatal SSRI exposure affects craniofacial form in multiple tissues and specifically at growth sites and centers of the skull.


Subject(s)
Citalopram , Craniofacial Abnormalities , Selective Serotonin Reuptake Inhibitors , Skull , Animals , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/adverse effects , Mice , Female , Pregnancy , Citalopram/pharmacology , Skull/drug effects , Craniofacial Abnormalities/chemically induced , Prenatal Exposure Delayed Effects/chemically induced , Disease Models, Animal , Male
12.
ACS Appl Mater Interfaces ; 16(30): 39035-39050, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39026394

ABSTRACT

Given the widespread clinical demand, addressing irregular cranial bone defects poses a significant challenge following surgical procedures and traumatic events. In situ-formed injectable hydrogels are attractive for irregular bone defects due to their ease of administration and the ability to incorporate ceramics, ions, and proteins into the hydrogel. In this study, a multifunctional hydrogel composed of oxidized sodium alginate (OSA)-grafted dopamine (DO), carboxymethyl chitosan (CMCS), calcium ions (Ca2+), nanohydroxyapatite (nHA), and magnesium oxide (MgO) (DOCMCHM) was prepared to address irregular cranial bone defects via dynamic Schiff base and chelation reactions. DOCMCHM hydrogel exhibits strong adhesion to wet tissues, self-healing properties, and antibacterial characteristics. Biological evaluations indicate that DOCMCHM hydrogel has good biocompatibility, in vivo degradability, and the ability to promote cell proliferation. Importantly, DOCMCHM hydrogel, containing MgO, promotes the expression of osteogenic protein markers COL-1, OCN, and RUNX2, and stimulates the formation of new blood vessels by upregulating CD31. This study could provide meaningful insights into ion therapy for the repair of cranial bone defects.


Subject(s)
Alginates , Anti-Bacterial Agents , Chitosan , Hydrogels , Skull , Hydrogels/chemistry , Hydrogels/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Chitosan/chemistry , Chitosan/analogs & derivatives , Chitosan/pharmacology , Animals , Alginates/chemistry , Skull/drug effects , Skull/pathology , Skull/diagnostic imaging , Skull/injuries , Magnesium Oxide/chemistry , Magnesium Oxide/pharmacology , Bone Regeneration/drug effects , Dopamine/chemistry , Dopamine/pharmacology , Durapatite/chemistry , Durapatite/pharmacology , Mice , Cell Proliferation/drug effects , Calcium/metabolism , Calcium/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Osteogenesis/drug effects , Staphylococcus aureus/drug effects
13.
Acta Cir Bras ; 39: e392824, 2024.
Article in English | MEDLINE | ID: mdl-39046039

ABSTRACT

PURPOSE: to evaluate biocompatibility and osteogenic potential of hydroxyapatite/alginate composite after its implantation on rat calvarian critical bone defect. METHODS: thirty adults male Wistar rats were randomly distributed into two groups: GHA - critical bone defect filled with hydroxyapatite/alginate composite granules (HA/Alg) and CG - critical bone defect without biomaterial; evaluated at biological points of 15, 45 and 120 days. RESULTS: the histomorphometrically analyses for GHA showed osteoid matrix deposition (OM) among the granules and towards the center of the defect in centripetal direction throughout the study, with evident new bone formation at 120 days, resulting in filling 4/5 of the initial bone defect. For CG, this finding was restricted to the edges of the bone margins and formation of connective tissue on the residual area was found in all biological points. Inflammatory response on GHA was chronic granulomatous type, discrete and regressive for all biological points. Throughout the study, the CG presented mononuclear inflammatory infiltrate diffuse and regressive. Histomorphometry analyses showed that OM percentage was evident for GHA group when compared to CG group in all analyzed periods (p > 0.05). CONCLUSIONS: the biomaterial evaluated at this study showed to be biocompatible, bioactive, osteoconductive and biodegradable synchronously with bone formation.


Subject(s)
Alginates , Biocompatible Materials , Bone Regeneration , Bone Substitutes , Durapatite , Materials Testing , Rats, Wistar , Animals , Male , Bone Regeneration/drug effects , Bone Regeneration/physiology , Alginates/pharmacology , Durapatite/pharmacology , Durapatite/therapeutic use , Biocompatible Materials/therapeutic use , Bone Substitutes/therapeutic use , Random Allocation , Osteogenesis/drug effects , Osteogenesis/physiology , Hexuronic Acids/pharmacology , Glucuronic Acid/pharmacology , Skull/surgery , Skull/drug effects , Time Factors , Rats , Reproducibility of Results
14.
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
15.
Cell Tissue Bank ; 25(3): 755-764, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38976150

ABSTRACT

Cryopreservation is a method adopted for storage of autologous skulls. Herein, this current research sought to explore the effects of different cryoprotectants on the biological characteristics of rat calvarial osteoblasts after cryopreservation. Neonatal Sprague-Dawley rats were selected and their skull tissues were isolated. The skull tissues were allocated into the refrigerating-3M, refrigerating-6M, M199-3M, M199-6M, povidone iodine-3M, and povidone iodine-6M groups according to the usage of cryoprotectants and treatment time (month) and the fresh group. Osteoblasts were isolated from skull tissues in each group through digestion. The histomorphology of the skull was evaluated by H&E staining and cell morphology was observed by microscopy. The viability, proliferation, apoptosis, and osteogenic activity of osteoblasts were assessed by trypan blue staining, MTT, flow cytometry, and alkaline phosphatase (ALP) staining. The skull histomorphology and osteoblast morphology were similar between the fresh and refrigerating groups. Osteoblast viability was weakened after cryopreservation. The longer the refrigeration time, the lower the number of living cells and the higher the apoptosis rate. However, cryopreservation using different cryoprotectants did not evidently affect osteoblast proliferation and ALP activity. Different cryoprotectants show no apparent effect on the osteogenic activity of rat calvarial osteoblasts after cryopreservation.


Subject(s)
Apoptosis , Cell Proliferation , Cell Survival , Cryopreservation , Cryoprotective Agents , Osteoblasts , Rats, Sprague-Dawley , Skull , Animals , Cryopreservation/methods , Osteoblasts/cytology , Osteoblasts/drug effects , Skull/cytology , Skull/drug effects , Cell Survival/drug effects , Cryoprotective Agents/pharmacology , Cell Proliferation/drug effects , Apoptosis/drug effects , Rats , Osteogenesis/drug effects , Cells, Cultured
16.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38928145

ABSTRACT

Polyurethane (PU) is a promising material for addressing challenges in bone grafting. This study was designed to enhance the bone grafting capabilities of PU by integrating hydroxyapatite (HAp), which is known for its osteoconductive and osteoinductive potential. Moreover, a uniform distribution of HAp in the porous structure of PU increased the effectiveness of bone grafts. PEG/APTES-modified scaffolds were prepared through self-foaming reactions. A uniform pore structure was generated during the spontaneous foaming reaction, and HAp was uniformly distributed in the PU structure (PU15HAp and PU30HAp) during foaming. Compared with the PU scaffolds, the HAp-modified PU scaffolds exhibited significantly greater protein absorption. Importantly, the effect of the HAp-modified PU scaffold on bone repair was tested in a rat calvarial defect model. The microstructure of the newly formed bone was analyzed with microcomputed tomography (µ-CT). Bone regeneration at the defect site was significantly greater in the HAp-modified PU scaffold group than in the PU group. This innovative HAp-modified PU scaffold improves current bone graft materials, providing a promising avenue for improved bone regeneration.


Subject(s)
Bone Regeneration , Durapatite , Polyurethanes , Skull , Tissue Scaffolds , Polyurethanes/chemistry , Animals , Durapatite/chemistry , Tissue Scaffolds/chemistry , Rats , Bone Regeneration/drug effects , Skull/drug effects , Skull/injuries , Skull/pathology , Skull/metabolism , Rats, Sprague-Dawley , X-Ray Microtomography , Male , Porosity , Bone Transplantation/methods
17.
Biomed Pharmacother ; 177: 117032, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38941894

ABSTRACT

In cell-based bone augmentation, transplanted cell dysfunction and apoptosis can occur due to oxidative stress caused by the overproduction of reactive oxygen species (ROS). Edaravone (EDA) is a potent free radical scavenger with potential medical applications. This study aimed to investigate the effect of controlling oxidative stress on bone regeneration using EDA. Bone marrow-derived cells were collected from 4-week-old rats, and EDA effects on cell viability and osteogenic differentiation were evaluated. Collagen gels containing PKH26-prelabeled cells were implanted into the calvarial defects of 12-week-old rats, followed by daily subcutaneous injections of normal saline or 500 µM EDA for 4 d. Bone formation was examined using micro-computed tomography and histological staining. Immunofluorescence staining was performed for markers of oxidative stress, macrophages, osteogenesis, and angiogenesis. EDA suppressed ROS production and hydrogen peroxide-induced apoptosis, recovering cell viability and osteoblast differentiation. EDA treatment in vivo increased new bone formation. EDA induced the transition of the macrophage population toward the M2 phenotype. The EDA group also exhibited stronger immunofluorescence for vascular endothelial growth factor and CD31. In addition, more PKH26-positive and PKH26-osteocalcin-double-positive cells were observed in the EDA group, indicating that transplanted cell survival was prolonged, and they differentiated into bone-forming cells. This could be attributed to oxidative stress suppression at the transplantation site by EDA. Collectively, local administration using EDA facilitates bone regeneration by improving the local environment and angiogenesis, prolonging survival, and enhancing the osteogenic capabilities of transplanted cells.


Subject(s)
Bone Regeneration , Cell Differentiation , Cell Survival , Edaravone , Osteogenesis , Oxidation-Reduction , Oxidative Stress , Rats, Sprague-Dawley , Reactive Oxygen Species , Animals , Bone Regeneration/drug effects , Edaravone/pharmacology , Osteogenesis/drug effects , Oxidative Stress/drug effects , Cell Differentiation/drug effects , Reactive Oxygen Species/metabolism , Rats , Cell Survival/drug effects , Male , Apoptosis/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Macrophages/drug effects , Macrophages/metabolism , Skull/drug effects , Cells, Cultured , Free Radical Scavengers/pharmacology , Free Radical Scavengers/administration & dosage
18.
PLoS One ; 19(6): e0304557, 2024.
Article in English | MEDLINE | ID: mdl-38941348

ABSTRACT

Prenatal alcohol exposure (PAE) causes cognitive impairment and a distinctive craniofacial dysmorphology, due in part to apoptotic losses of the pluripotent cranial neural crest cells (CNCs) that form facial bones and cartilage. We previously reported that PAE rapidly represses expression of >70 ribosomal proteins (padj = 10-E47). Ribosome dysbiogenesis causes nucleolar stress and activates p53-MDM2-mediated apoptosis. Using primary avian CNCs and the murine CNC line O9-1, we tested whether nucleolar stress and p53-MDM2 signaling mediates this apoptosis. We further tested whether haploinsufficiency in genes that govern ribosome biogenesis, using a blocking morpholino approach, synergizes with alcohol to worsen craniofacial outcomes in a zebrafish model. In both avian and murine CNCs, pharmacologically relevant alcohol exposure (20mM, 2hr) causes the dissolution of nucleolar structures and the loss of rRNA synthesis; this nucleolar stress persisted for 18-24hr. This was followed by reduced proliferation, stabilization of nuclear p53, and apoptosis that was prevented by overexpression of MDM2 or dominant-negative p53. In zebrafish embryos, low-dose alcohol or morpholinos directed against ribosomal proteins Rpl5a, Rpl11, and Rps3a, the Tcof homolog Nolc1, or mdm2 separately caused modest craniofacial malformations, whereas these blocking morpholinos synergized with low-dose alcohol to reduce and even eliminate facial elements. Similar results were obtained using a small molecule inhibitor of RNA Polymerase 1, CX5461, whereas p53-blocking morpholinos normalized craniofacial outcomes under high-dose alcohol. Transcriptome analysis affirmed that alcohol suppressed the expression of >150 genes essential for ribosome biogenesis. We conclude that alcohol causes the apoptosis of CNCs, at least in part, by suppressing ribosome biogenesis and invoking a nucleolar stress that initiates their p53-MDM2 mediated apoptosis. We further note that the facial deficits that typify PAE and some ribosomopathies share features including reduced philtrum, upper lip, and epicanthal distance, suggesting the facial deficits of PAE represent, in part, a ribosomopathy.


Subject(s)
Apoptosis , Ethanol , Neural Crest , Ribosomes , Tumor Suppressor Protein p53 , Zebrafish , Animals , Neural Crest/metabolism , Neural Crest/drug effects , Ribosomes/metabolism , Ribosomes/drug effects , Ethanol/toxicity , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Mice , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Cell Nucleolus/metabolism , Cell Nucleolus/drug effects , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Skull/pathology , Skull/metabolism , Skull/drug effects , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
19.
ACS Biomater Sci Eng ; 10(7): 4452-4462, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38875708

ABSTRACT

Mg-based biodegradable metallic implants are gaining increased attraction for applications in orthopedics and dentistry. However, their current applications are hampered by their high rate of corrosion, degradation, and rapid release of ions and gas bubbles into the physiological medium. The aim of the present study is to investigate the osteogenic and angiogenic potential of coated Mg-based implants in a sheep cranial defect model. Although their osteogenic potential was studied to some extent, their potential to regenerate vascularized bone formation was not studied in detail. We have studied the potential of magnesium-calcium (MgCa)-based alloys modified with zinc (Zn)- or gallium (Ga)-doped calcium phosphate (CaP) coatings as a strategy to control their degradation rate while enhancing bone regeneration capacity. MgCa and its implants with CaP coatings (MgCa/CaP) as undoped or as doped with Zn or Ga (MgCa/CaP + Zn and MgCa/CaP + Ga, respectively) were implanted in bone defects created in the sheep cranium. MgCa implants degraded faster than the others at 4 weeks postop and the weight loss was ca. 50%, while it was ca. 15% for MgCa/CaP and <10% in the presence of Zn and Ga with CaP coating. Scanning electron microscopy (SEM) analysis of the implant surfaces also revealed that the MgCa implants had the largest degree of structural breakdown of all the groups. Radiological evaluation revealed that surface modification with CaP to the MgCa implants induced better bone regeneration within the defects as well as the enhancement of bone-implant surface integration. Bone volume (%) within the defect was ca. 25% in the case of MgCa/CaP + Ga, while it was around 15% for undoped MgCa group upon micro-CT evaluation. This >1.5-fold increase in bone regeneration for MgCa/CaP + Ga implant was also observed in the histopathological examination of the H&E- and Masson's trichrome-stained sections. Immunohistochemical analysis of the bone regeneration (antiosteopontin) and neovascularization (anti-CD31) at the defect sites revealed >2-fold increase in the expression of the markers in both Ga- and Zn-doped, CaP-coated implants. Zn-doped implants further presented low inflammatory reaction, notable bone regeneration, and neovascularization among all the implant groups. These findings indicated that Ga- and Zn-doped CaP coating is an important strategy to control the degradation rate as well as to achieve enhanced bone regeneration capacity of the implants made of Mg-based alloys.


Subject(s)
Alloys , Calcium Phosphates , Coated Materials, Biocompatible , Gallium , Magnesium , Osteogenesis , Skull , Zinc , Animals , Zinc/chemistry , Zinc/pharmacology , Sheep , Skull/drug effects , Skull/pathology , Skull/injuries , Osteogenesis/drug effects , Magnesium/pharmacology , Gallium/chemistry , Gallium/pharmacology , Alloys/chemistry , Alloys/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Bone Regeneration/drug effects , Calcium/metabolism , Absorbable Implants
20.
Biomater Adv ; 161: 213892, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795472

ABSTRACT

Guided bone regeneration (GBR) stands as an essential modality for craniomaxillofacial bone defect repair, yet challenges like mechanical weakness, inappropriate degradability, limited bioactivity, and intricate manufacturing of GBR membranes hindered the clinical efficacy. Herein, we developed a Janus bacterial cellulose(BC)/MXene membrane through a facile vacuum filtration and etching strategy. This Janus membrane displayed an asymmetric bilayer structure with interfacial compatibility, where the dense layer impeded cell invasion and the porous layer maintained stable space for osteogenesis. Incorporating BC with Ti3C2Tx MXene significantly enhanced the mechanical robustness and flexibility of the material, enabling clinical operability and lasting GBR membrane supports. It also contributed to a suitable biodegradation rate, which aligned with the long-term bone repair period. After demonstrating the desirable biocompatibility, barrier role, and osteogenic capability in vitro, the membrane's regenerative potential was also confirmed in a rat cranial defect model. The excellent bone repair performance could be attributed to the osteogenic capability of MXene nanosheets, the morphological cues of the porous layer, as well as the long-lasting, stable regeneration space provided by the GBR membrane. Thus, our work presented a facile, robust, long-lasting, and biodegradable BC/MXene GBR membrane, offering a practical solution to craniomaxillofacial bone defect repair.


Subject(s)
Bone Regeneration , Cellulose , Guided Tissue Regeneration , Osteogenesis , Titanium , Bone Regeneration/drug effects , Cellulose/chemistry , Animals , Rats , Titanium/chemistry , Titanium/pharmacology , Guided Tissue Regeneration/methods , Osteogenesis/drug effects , Membranes, Artificial , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Rats, Sprague-Dawley , Humans , Porosity , Skull/surgery , Skull/drug effects , Skull/injuries
SELECTION OF CITATIONS
SEARCH DETAIL