Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.035
Filtrar
1.
Acta Biomater ; 186: 411-423, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39089349

RESUMEN

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.


Asunto(s)
Regeneración Ósea , Membrana Celular , Animales , Ratones , Regeneración Ósea/efectos de los fármacos , Membrana Celular/metabolismo , Osteoblastos/metabolismo , Osteoblastos/citología , Cráneo/patología , Cráneo/lesiones , Condrocitos/metabolismo , Condrocitos/citología , Línea Celular , Osteogénesis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos
2.
Int J Nanomedicine ; 19: 8309-8336, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39161358

RESUMEN

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.


Asunto(s)
Regeneración Ósea , Cobre , Grafito , Hidrogeles , Ratas Sprague-Dawley , Cráneo , Ingeniería de Tejidos , Andamios del Tejido , Animales , Regeneración Ósea/efectos de los fármacos , Andamios del Tejido/química , Cobre/química , Cobre/farmacología , Grafito/química , Hidrogeles/química , Hidrogeles/farmacología , Cráneo/efectos de los fármacos , Cráneo/lesiones , Ratas , Ratones , Ingeniería de Tejidos/métodos , Osteogénesis/efectos de los fármacos , Ratones Endogámicos BALB C , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Masculino , Nanopartículas del Metal/química , Nanoestructuras/química , Gelatina/química , Células RAW 264.7
3.
Stem Cells Transl Med ; 13(8): 791-802, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-38986535

RESUMEN

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.


Asunto(s)
Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Cráneo , Animales , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Ratones , Cráneo/metabolismo , Cráneo/lesiones , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Regeneración Ósea/fisiología
4.
Biomater Sci ; 12(16): 4226-4241, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38984522

RESUMEN

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.


Asunto(s)
Regeneración Ósea , Dentina , Cráneo , Animales , Regeneración Ósea/efectos de los fármacos , Cráneo/lesiones , Cráneo/patología , Cráneo/diagnóstico por imagen , Cráneo/efectos de los fármacos , Ratas , Dentina/química , Ratas Sprague-Dawley , Membranas Artificiales , Masculino , Cicatrización de Heridas/efectos de los fármacos , Microtomografía por Rayos X , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Andamios del Tejido/química , Osteogénesis/efectos de los fármacos
5.
ACS Appl Mater Interfaces ; 16(30): 39035-39050, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39026394

RESUMEN

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.


Asunto(s)
Alginatos , Antibacterianos , Quitosano , Hidrogeles , Cráneo , Hidrogeles/química , Hidrogeles/farmacología , Antibacterianos/química , Antibacterianos/farmacología , Quitosano/química , Quitosano/análogos & derivados , Quitosano/farmacología , Animales , Alginatos/química , Cráneo/efectos de los fármacos , Cráneo/patología , Cráneo/diagnóstico por imagen , Cráneo/lesiones , Óxido de Magnesio/química , Óxido de Magnesio/farmacología , Regeneración Ósea/efectos de los fármacos , Dopamina/química , Dopamina/farmacología , Durapatita/química , Durapatita/farmacología , Ratones , Proliferación Celular/efectos de los fármacos , Calcio/metabolismo , Calcio/química , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Osteogénesis/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos
6.
Bull Exp Biol Med ; 177(1): 155-161, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38963597

RESUMEN

Experimental model of resection craniotomy with subsequent reconstruction of the defect with a polymer implant enables comprehensive assessment of functional and ultrastructural changes during replacement of the damaged tissue. Reconstruction of a skull defect was accompanied by transient motor disturbance in the acute period and did not cause functional disorders and neurological deficits in a delayed period. Histological examination of osteal and brain tissue revealed no pathological reactions that could be associated with the response to the chemical components of the implant.


Asunto(s)
Benzofenonas , Craneotomía , Polietilenglicoles , Polímeros , Cráneo , Polímeros/química , Animales , Cráneo/cirugía , Cráneo/lesiones , Cráneo/diagnóstico por imagen , Polietilenglicoles/química , Craneotomía/métodos , Ratas , Masculino , Procedimientos de Cirugía Plástica/métodos , Cetonas/química , Materiales Biocompatibles/química , Encéfalo/cirugía , Ratas Wistar
7.
Adv Healthc Mater ; 13(23): e2401031, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38850118

RESUMEN

Mobilizing endogenous progenitor cells to repair damaged tissue in situ has the potential to revolutionize the field of regenerative medicine, while the early establishment of a vascular network will ensure survival of newly generated tissue. In this study, a gene-activated scaffold containing a stromal derived factor 1α plasmid (pSDF1α), a pro-angiogenic gene that is also thought to be involved in the recruitment of mesenchymal stromal cells (MSCs) to sites of injury is described. It is shown that over-expression of SDF1α protein enhanced MSC recruitment and induced vessel-like structure formation by endothelial cells in vitro. When implanted subcutaneously, transcriptomic analysis reveals that endogenous MSCs are recruited and significant angiogenesis is stimulated. Just 1-week after implantation into a calvarial critical-sized bone defect, pSDF1α-activated scaffolds are recruited MSCs and rapidly activate angiogenic and osteogenic programs, upregulating Runx2, Dlx5, and Sp7. At the same time-point, pVEGF-activated scaffolds are recruited a variety of cell types, activating endochondral ossification. The early response induced by both scaffolds leads to complete bridging of the critical-sized bone defects within 4-weeks. The versatile cell-free gene-activated scaffold described in this study is capable of harnessing and enhancing the body's own regenerative capacity and has immense potential in a myriad of applications.


Asunto(s)
Regeneración Ósea , Quimiocina CXCL12 , Células Madre Mesenquimatosas , Neovascularización Fisiológica , Osteogénesis , Andamios del Tejido , Animales , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Andamios del Tejido/química , Regeneración Ósea/fisiología , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/genética , Osteogénesis/fisiología , Ratones , Humanos , Cráneo/lesiones , Cráneo/patología , Cráneo/metabolismo , Angiogénesis
8.
ACS Biomater Sci Eng ; 10(7): 4452-4462, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38875708

RESUMEN

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.


Asunto(s)
Aleaciones , Fosfatos de Calcio , Materiales Biocompatibles Revestidos , Galio , Magnesio , Osteogénesis , Cráneo , Zinc , Animales , Zinc/química , Zinc/farmacología , Ovinos , Cráneo/efectos de los fármacos , Cráneo/patología , Cráneo/lesiones , Osteogénesis/efectos de los fármacos , Magnesio/farmacología , Galio/química , Galio/farmacología , Aleaciones/química , Aleaciones/farmacología , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Fosfatos de Calcio/química , Fosfatos de Calcio/farmacología , Regeneración Ósea/efectos de los fármacos , Calcio/metabolismo , Implantes Absorbibles
9.
Int J Mol Sci ; 25(12)2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38928145

RESUMEN

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.


Asunto(s)
Regeneración Ósea , Durapatita , Poliuretanos , Cráneo , Andamios del Tejido , Poliuretanos/química , Animales , Durapatita/química , Andamios del Tejido/química , Ratas , Regeneración Ósea/efectos de los fármacos , Cráneo/efectos de los fármacos , Cráneo/lesiones , Cráneo/patología , Cráneo/metabolismo , Ratas Sprague-Dawley , Microtomografía por Rayos X , Masculino , Porosidad , Trasplante Óseo/métodos
10.
J Vis Exp ; (206)2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38709029

RESUMEN

Mild traumatic brain injury is a clinically highly heterogeneous neurological disorder. Highly reproducible traumatic brain injury (TBI) animal models with well-defined pathologies are urgently needed for studying the mechanisms of neuropathology after mild TBI and testing therapeutics. Replicating the entire sequelae of TBI in animal models has proven to be a challenge. Therefore, the availability of multiple animal models of TBI is necessary to account for the diverse aspects and severities seen in TBI patients. CHI is one of the most common methods for fabricating rodent models of rmTBI. However, this method is susceptible to many factors, including the impact method used, the thickness and shape of the skull bone, animal apnea, and the type of head support and immobilization utilized. The aim of this protocol is to demonstrate a combination of the thinned-skull window and fluid percussion injury (FPI) methods to produce a precise mouse model of CHI-associated rmTBI. The primary objective of this protocol is to minimize factors that could impact the accuracy and consistency of CHI and FPI modeling, including skull bone thickness, shape, and head support. By utilizing a thinned-skull window method, potential inflammation due to craniotomy and FPI is minimized, resulting in an improved mouse model that replicates the clinical features observed in patients with mild TBI. Results from behavior and histological analysis using hematoxylin and eosin (HE) staining suggest that rmTBI can lead to a cumulative injury that produces changes in both behavior and gross morphology of the brain. Overall, the modified CHI-associated rmTBI presents a useful tool for researchers to explore the underlying mechanisms that contribute to focal and diffuse pathophysiological changes in rmTBI.


Asunto(s)
Conmoción Encefálica , Modelos Animales de Enfermedad , Cráneo , Animales , Ratones , Conmoción Encefálica/patología , Cráneo/patología , Cráneo/lesiones , Cráneo/cirugía , Masculino , Percusión/métodos , Lesiones Traumáticas del Encéfalo/patología
11.
Biomater Adv ; 161: 213892, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38795472

RESUMEN

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.


Asunto(s)
Regeneración Ósea , Celulosa , Regeneración Tisular Dirigida , Osteogénesis , Titanio , Regeneración Ósea/efectos de los fármacos , Celulosa/química , Animales , Ratas , Titanio/química , Titanio/farmacología , Regeneración Tisular Dirigida/métodos , Osteogénesis/efectos de los fármacos , Membranas Artificiales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Ratas Sprague-Dawley , Humanos , Porosidad , Cráneo/cirugía , Cráneo/efectos de los fármacos , Cráneo/lesiones
12.
Int J Biol Macromol ; 270(Pt 2): 132419, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38759859

RESUMEN

Bacterial infection is a serious challenge in the treatment of open bone defects, and reliance on antibiotic therapy may contribute to the emergence of drug-resistant bacteria. To solve this problem, this study developed a mineralized hydrogel (PVA-Ag-PHA) with excellent antibacterial properties and osteogenic capabilities. Silver nanoparticles (CNC/TA@AgNPs) were greenly synthesized using natural macromolecular cellulose nanocrystals (CNC) and plant polyphenolic tannins (TA) as stabilizers and reducing agents respectively, and then introduced into polyvinyl alcohol (PVA) and polydopamine-modified hydroxyapatite (PDA@HAP) hydrogel. The experimental results indicate that the PVA-Ag-PHA hydrogel, benefiting from the excellent antibacterial properties of CNC/TA@AgNPs, can not only eliminate Staphylococcus aureus and Escherichia coli, but also maintain a sustained sterile environment. At the same time, the HAP modified by PDA is uniformly dispersed within the hydrogel, thus releasing and maintaining stable concentrations of Ca2+ and PO43- ions in the local environment. The porous structure of the hydrogel with excellent biocompatibility creates a suitable bioactive environment that facilitates cell adhesion and bone regeneration. The experimental results in the rat critical-sized calvarial defect model indicate that the PVA-Ag-PHA hydrogel can effectively accelerate the bone healing process. Thus, this mussel-inspired hydrogel with antibacterial properties provides a feasible solution for the repair of open bone defects, demonstrating the considerable potential for diverse applications in bone repair.


Asunto(s)
Regeneración Ósea , Celulosa , Hidrogeles , Nanopartículas del Metal , Plata , Cráneo , Taninos , Plata/química , Plata/farmacología , Animales , Regeneración Ósea/efectos de los fármacos , Celulosa/química , Celulosa/farmacología , Nanopartículas del Metal/química , Ratas , Hidrogeles/química , Hidrogeles/farmacología , Cráneo/efectos de los fármacos , Cráneo/lesiones , Taninos/química , Taninos/farmacología , Bivalvos/química , Antibacterianos/farmacología , Antibacterianos/química , Alcohol Polivinílico/química , Staphylococcus aureus/efectos de los fármacos , Durapatita/química , Durapatita/farmacología , Ratas Sprague-Dawley , Escherichia coli/efectos de los fármacos
13.
Int J Biol Macromol ; 270(Pt 1): 132361, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38750857

RESUMEN

Critical-sized bone defects are a major challenge in reconstructive bone surgery and usually fail to be treated due to limited remaining bone quality and extensive healing time. The combination of 3D-printed scaffolds and bioactive materials is a promising approach for bone tissue regeneration. In this study, 3D-printed alkaline-treated polycaprolactone scaffolds (M-PCL) were fabricated and integrated with tragacanth gum- 45S5 bioactive glass (TG-BG) to treat critical-sized calvarial bone defects in female adult Wistar rats. After a healing period of four and eight weeks, the new bone of blank, M-PCL, and M-PCL/TG-BG groups were harvested and assessed. Micro-computed tomography, histological, biochemical, and biomechanical analyses, gene expression, and bone matrix formation were used to assess bone regeneration. The micro-computed tomography results showed that the M-PCL/TG-BG scaffolds not only induced bone tissue formation within the bone defect but also increased BMD and BV/TV compared to blank and M-PCL groups. According to the histological analysis, there was no evidence of bony union in the calvarial defect regions of blank groups, while in M-PCL/TG-BG groups bony integration and repair were observed. The M-PCL/TG-BG scaffolds promoted the Runx2 and collagen type I expression as compared with blank and M-PCL groups. Besides, the bone regeneration in M-PCL/TG-BG groups correlated with TG-BG incorporation. Moreover, the use of M-PCL/TG-BG scaffolds promoted the biomechanical properties in the bone remodeling process. These data demonstrated that the M-PCL/TG-BG scaffolds serve as a highly promising platform for the development of bone grafts, supporting bone regeneration with bone matrix formation, and osteogenic features. Our results exhibited that the 3D-printed M-PCL/TG-BG scaffolds are a promising strategy for successful bone regeneration.


Asunto(s)
Regeneración Ósea , Vidrio , Osteogénesis , Poliésteres , Impresión Tridimensional , Ratas Wistar , Cráneo , Andamios del Tejido , Animales , Poliésteres/química , Andamios del Tejido/química , Ratas , Regeneración Ósea/efectos de los fármacos , Cráneo/efectos de los fármacos , Cráneo/patología , Cráneo/lesiones , Cráneo/diagnóstico por imagen , Osteogénesis/efectos de los fármacos , Femenino , Vidrio/química , Tragacanto/química , Microtomografía por Rayos X , Ingeniería de Tejidos/métodos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología
14.
J Forensic Sci ; 69(4): 1171-1182, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38798041

RESUMEN

Skeletal evidence usually constitutes the only source of information to interpret lesion patterns that help to clarify the circumstances surrounding death. The examination and interpretation of bone trauma are essential to the application and utility of anthropology as a forensic science. When discussing the effect of gunshot wounds in bone, it becomes imperative to differentiate between short and long-distance injuries based on clear, distinct, and observable signs. To contribute to the debate, our focus is directed toward the external analysis of the so-called circumferential delamination defect (CDD) as an observable proxy for close-range shooting (≤30 cm) and contact gunshot wounds in the skull. In the context of known extrajudicial killings, in which the perpetrators used short 9 × 19 FMJ ammunition in a close-range shooting, instances of CDD have been documented. Empirical evidence reinforcing the causal relationship between CDD and close-range shootings is presented. Elements' characteristics of firearm residues were also found in remains buried for up to 30 years. Primarily, this work shows that the concentrations of gunshot residues (Pb, Ba, and Sb) resemble those observed in fresh corpses with the same gunshot wound (GSW). Moreover, the correlation observed between CDD and gunshot residues, where the likelihood of CDD increases the closer to the head and the more perpendicular the shot angle is, reinforces CDD as a pivotal discriminatory factor in the skeletal evidence of short-range or contact shot. This research contributes to the field of forensic anthropology by providing fundamental insights into the etiology of CDD and its practical application.


Asunto(s)
Armas de Fuego , Balística Forense , Traumatismos Penetrantes de la Cabeza , Heridas por Arma de Fuego , Humanos , Heridas por Arma de Fuego/patología , Masculino , Traumatismos Penetrantes de la Cabeza/patología , Bario/análisis , Plomo/análisis , Adulto , Persona de Mediana Edad , Homicidio , Cráneo/lesiones , Cráneo/patología , Ciclohexanonas
15.
Colloids Surf B Biointerfaces ; 239: 113971, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38759296

RESUMEN

The optimal material for repairing skull defects should exhibit outstanding biocompatibility and mechanical properties. Specifically, hydrogel scaffolds that emulate the microenvironment of the native bone extracellular matrix play a vital role in promoting osteoblast adhesion, proliferation, and differentiation, thereby yielding superior outcomes in skull reconstruction. In this study, a composite network hydrogel comprising sodium alginate (SA), epigallocatechin gallate (EGCG), and zinc ions (Zn2+) was developed to establish an ideal osteogenic microenvironment for bone regeneration. Initially, physical entanglement and hydrogen bonding between SA and EGCG resulted in the formation of a primary network hydrogel known as SA-EGCG. Subsequently, the inclusion of Zn2+ facilitated the creation of a composite network hydrogels named SA-EGCG-Zn2+ via dynamic coordination bonds with SA and EGCG. The engineered SA-EGCG2 %-Zn2+ hydrogels offered an environment mimicking the native extracellular matrix (ECM). Moreover, the sustained release of Zn2+ from the hydrogel effectively enhanced cell adhesion, promoted proliferation, and stimulated osteoblast differentiation. In vitro experiments have shown that SA-EGCG2 %-Zn2+ hydrogels greatly enhance the attachment and growth of osteoblast precursor cells (MC3T3-E1), while also increasing the expression of genes related to osteogenesis in these cells. Additionally, in vivo studies have confirmed that SA-EGCG2 %-Zn2+ hydrogels promote new bone formation and accelerate the regeneration of bone in situ, indicating promising applications in the realm of bone tissue engineering.


Asunto(s)
Alginatos , Catequina , Proliferación Celular , Hidrogeles , Cráneo , Andamios del Tejido , Zinc , Zinc/química , Zinc/farmacología , Alginatos/química , Alginatos/farmacología , Catequina/química , Catequina/análogos & derivados , Catequina/farmacología , Cráneo/efectos de los fármacos , Cráneo/lesiones , Cráneo/patología , Animales , Ratones , Hidrogeles/química , Hidrogeles/farmacología , Andamios del Tejido/química , Proliferación Celular/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/metabolismo , Diferenciación Celular/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Regeneración Ósea/efectos de los fármacos , Adhesión Celular/efectos de los fármacos
16.
Biomacromolecules ; 25(6): 3784-3794, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38743836

RESUMEN

The effective regeneration of large bone defects via bone tissue engineering is challenging due to the difficulty in creating an osteogenic microenvironment. Inspired by the fibrillar architecture of the natural extracellular matrix, we developed a nanoscale bioengineering strategy to produce bone fibril-like composite scaffolds with enhanced osteogenic capability. To activate the surface for biofunctionalization, self-adaptive ridge-like nanolamellae were constructed on poly(ε-caprolactone) (PCL) electrospinning scaffolds via surface-directed epitaxial crystallization. This unique nanotopography with a markedly increased specific surface area offered abundant nucleation sites for Ca2+ recruitment, leading to a 5-fold greater deposition weight of hydroxyapatite than that of the pristine PCL scaffold under stimulated physiological conditions. Bone marrow mesenchymal stem cells (BMSCs) cultured on bone fibril-like scaffolds exhibited enhanced adhesion, proliferation, and osteogenic differentiation in vitro. In a rat calvarial defect model, the bone fibril-like scaffold significantly accelerated bone regeneration, as evidenced by micro-CT, histological histological and immunofluorescence staining. This work provides the way for recapitulating the osteogenic microenvironment in tissue-engineered scaffolds for bone repair.


Asunto(s)
Regeneración Ósea , Células Madre Mesenquimatosas , Osteogénesis , Poliésteres , Ingeniería de Tejidos , Andamios del Tejido , Animales , Andamios del Tejido/química , Ratas , Regeneración Ósea/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Ingeniería de Tejidos/métodos , Poliésteres/química , Diferenciación Celular , Ratas Sprague-Dawley , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Células Cultivadas , Proliferación Celular , Cráneo/lesiones , Cráneo/patología , Durapatita/química , Durapatita/farmacología
17.
Biomed Mater ; 19(3)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38626780

RESUMEN

Wool derived keratin, due to its demonstrated ability to promote bone formation, has been suggested as a potential bioactive material for implant surfaces. The aim of this study was to assess the effects of keratin-coated titanium on osteoblast functionin vitroand bone healingin vivo. Keratin-coated titanium surfaces were fabricated via solvent casting and molecular grafting. The effect of these surfaces on the attachment, osteogenic gene, and osteogenic protein expression of MG-63 osteoblast-like cells were quantifiedin vitro. The effect of these keratin-modified surfaces on bone healing over three weeks using an intraosseous calvaria defect was assessed in rodents. Keratin coating did not affect MG-63 proliferation or viability, but enhanced osteopontin, osteocalcin and bone morphogenetic expressionin vitro. Histological analysis of recovered calvaria specimens showed osseous defects covered with keratin-coated titanium had a higher percentage of new bone area two weeks after implantation compared to that in defects covered with titanium alone. The keratin-coated surfaces were biocompatible and stimulated osteogenic expression in adherent MG-63 osteoblasts. Furthermore, a pilot preclinical study in rodents suggested keratin may stimulate earlier intraosseous calvaria bone healing.


Asunto(s)
Regeneración Ósea , Proliferación Celular , Materiales Biocompatibles Revestidos , Queratinas , Osteoblastos , Osteogénesis , Cráneo , Titanio , Titanio/química , Osteoblastos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/metabolismo , Regeneración Ósea/efectos de los fármacos , Animales , Queratinas/química , Queratinas/metabolismo , Humanos , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Proliferación Celular/efectos de los fármacos , Cráneo/efectos de los fármacos , Cráneo/lesiones , Osteogénesis/efectos de los fármacos , Ratas , Propiedades de Superficie , Masculino , Línea Celular , Adhesión Celular/efectos de los fármacos , Ensayo de Materiales , Supervivencia Celular/efectos de los fármacos , Ratas Sprague-Dawley
18.
J Control Release ; 370: 277-286, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38679161

RESUMEN

Addressing bone defects represents a significant challenge to public health. Localized delivery of growth factor has emerged as promising approach for bone regeneration. However, the clinical application of Platelet-Derived Growth Factor (PDGF) is hindered by its high cost and short half-life. In this work, we introduce the application of PDGF-mimicking peptide (PMP1) hydrogels for calvarial defect restoration, showcasing their remarkable effectiveness. Through osteogenic differentiation assays and q-PCR analyses, we demonstrate PMP1's substantial capacity to enhance osteogenic differentiation of bone marrow mesenchymal stem cell (BMSC), leading to increased expression of crucial osteogenic genes. Further molecular mechanistic investigations reveal PMP1's activation of the PI3K-AKT-mTOR signaling pathway, a key element of its osteogenic effect. In vivo experiments utilizing a rat calvaria critical-sized defect model underscore the hydrogels' exceptional ability to accelerate new bone formation, thereby significantly advancing the restoration of calvaria defects. This research provides a promising bioactive material for bone tissue regeneration.


Asunto(s)
Becaplermina , Regeneración Ósea , Diferenciación Celular , Hidrogeles , Células Madre Mesenquimatosas , Osteogénesis , Ratas Sprague-Dawley , Cráneo , Animales , Hidrogeles/química , Cráneo/efectos de los fármacos , Cráneo/lesiones , Osteogénesis/efectos de los fármacos , Becaplermina/administración & dosificación , Regeneración Ósea/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Masculino , Péptidos/química , Péptidos/administración & dosificación , Péptidos/farmacología , Células Cultivadas , Ratas
19.
Leg Med (Tokyo) ; 69: 102443, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38569417

RESUMEN

Depressed skull fractures occur when broken bones displace inward, meaning that a portion of the outer table of the fracture line lies below the normal anatomical position of the inner table. They typically result from force trauma, when the skull is struck by an object with a moderately large amount of kinetic energy but a small surface area, or when an object with a large amount of kinetic energy impacts only a small area of the skull. In the present case, a depressed fracture of the frontal bone was detected at the autopsy of a 52-year-old man who, according to the belated confession of the assailant, was kicked in the head. The assailant was wearing sneakers. Could such a fracture be caused "just" by a kick? In this case it was possible due to an extraordinarily thin cranial vault (0.2 cm frontal, 0.3 cm occipital), which allowed the fractures to occur from a kinetic force that might not have been sufficient with a normal cranial vault thickness. An important role in the forensic analysis of the case was played by the 3D CT reconstruction.


Asunto(s)
Imagenología Tridimensional , Fractura Craneal Deprimida , Tomografía Computarizada por Rayos X , Humanos , Masculino , Persona de Mediana Edad , Fractura Craneal Deprimida/diagnóstico por imagen , Fractura Craneal Deprimida/patología , Autopsia/métodos , Cráneo/diagnóstico por imagen , Cráneo/lesiones , Cráneo/patología , Fracturas Craneales/diagnóstico por imagen , Fracturas Craneales/patología , Patologia Forense/métodos
20.
Leg Med (Tokyo) ; 69: 102445, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38640873

RESUMEN

A smoothbore musket firing a round ball was the primary weapon of the infantry from the 16th to mid 19th century. Musket ball injuries are thus relatively common when archaeological remains of battlefield victims from that period are studied. Several experimental studies have focused on terminal ballistics of a musket ball. In addition, there is a good supply of historical records directly from the battlefield and military hospitals. Studies and historical records have both concluded that head injuries are among the most lethal types of musket ball damage. In this study we utilized modern day research methods, including Synbone ballistic skull phantoms and computed tomography (CT) imaging, to examine more closely the head injuries and tissue damage caused by a musket ball. We were especially interested to observe how different musket ball velocities and shooting distances would influence bone and soft tissue defects. Our experiments clearly demonstrated that musket ball was a lethal projectile even from a longer distance. Already at low velocities, the musket ball perforated through the skull. Velocity also influenced the appearance of entrance and exit wounds. CT imaging provided us with a three-dimensional view of the wound channel, skull fragments and lead remnants inside the skull phantom. According to our findings, musket ball velocity influenced defect size and cavitation. In addition, velocity influenced the size and distribution of skull fragments and lead remnants in the wound channel. Combining all these aspects could aid us in studies of archaeological musket ball victims. In particular, they could help us to estimate the shooting distance and shed light on the potential course of events in the battlefield.


Asunto(s)
Balística Forense , Tomografía Computarizada por Rayos X , Humanos , Balística Forense/métodos , Heridas por Arma de Fuego/diagnóstico por imagen , Cráneo/diagnóstico por imagen , Cráneo/lesiones , Traumatismos Penetrantes de la Cabeza/diagnóstico por imagen , Traumatismos Penetrantes de la Cabeza/historia , Traumatismos Penetrantes de la Cabeza/patología , Armas de Fuego , Fantasmas de Imagen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA