Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.125
Filter
1.
Dis Model Mech ; 17(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38967226

ABSTRACT

Robinow syndrome is a rare disease caused by variants of seven WNT pathway genes. Craniofacial features include widening of the nasal bridge and jaw hypoplasia. We used the chicken embryo to test whether two missense human FZD2 variants (1301G>T, p.Gly434Val; 425C>T, p.Pro142Lys) were sufficient to change frontonasal mass development. In vivo, the overexpression of retroviruses with wild-type or variant human FZD2 inhibited upper beak ossification. In primary cultures, wild-type and variant human FZD2 significantly inhibited chondrogenesis, with the 425C>T variant significantly decreasing activity of a SOX9 luciferase reporter compared to that for the wild type or 1301G>T. Both variants also increased nuclear shuttling of ß-catenin (CTNNB1) and increased the expression of TWIST1, which are inhibitory to chondrogenesis. In canonical WNT luciferase assays using frontonasal mass cells, the variants had dominant-negative effects on wild-type FZD2. In non-canonical assays, the 425C>T variant failed to activate the reporter above control levels and was unresponsive to exogenous WNT5A. This is the first single amino acid change to selectively alter ligand binding in a FZD receptor. Therefore, FZD2 missense variants are pathogenic and could lead to the altered craniofacial morphogenesis seen in Robinow syndrome.


Subject(s)
Chondrogenesis , Craniofacial Abnormalities , Frizzled Receptors , Animals , Humans , Chick Embryo , Chondrogenesis/genetics , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , beta Catenin/metabolism , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics , Beak , Skull/pathology , Skull/embryology , Cell Nucleus/metabolism , Wnt Signaling Pathway , Dwarfism , Urogenital Abnormalities , Limb Deformities, Congenital
2.
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
3.
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
4.
ACS Biomater Sci Eng ; 10(7): 4297-4310, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38900847

ABSTRACT

Interfaces between AISI 304 stainless steel screws and cranial bone were investigated after long-term implantation lasting for 42 years. Samples containing the interface regions were analyzed using state-of-the-art analytical techniques including secondary ion mass, Fourier-transform infrared, Raman, and X-ray photoelectron spectroscopies. Local samples for scanning transmission electron microscopy were cut from the interface regions using the focused ion beam technique. A chemical composition across the interface was recorded in length scales covering micrometric and nanometric resolutions and relevant differences were found between peri-implant and the distant cranial bone, indicating generally younger bone tissue in the peri-implant area. Furthermore, the energy dispersive spectroscopy revealed an 80 nm thick steel surface layer enriched by oxygen suggesting that the AISI 304 material undergoes a corrosion attack. The attack is associated with transport of metallic ions, namely, ferrous and ferric iron, into the bone layer adjacent to the implant. The results comply with an anticipated interplay between released iron ions and osteoclast proliferation. The interplay gives rise to an autocatalytic process in which the iron ions stimulate the osteoclast activity while a formation of fresh bone resorption sites boosts the corrosion process through interactions between acidic osteoclast extracellular compartments and the implant surface. The autocatalytic process thus may account for an accelerated turnover of the peri-implant bone.


Subject(s)
Bone Screws , Skull , Stainless Steel , Bone Screws/adverse effects , Stainless Steel/chemistry , Humans , Corrosion , Skull/pathology , Spectroscopy, Fourier Transform Infrared , Bone-Implant Interface , Surface Properties , Photoelectron Spectroscopy , Spectrum Analysis, Raman , Iron/chemistry
5.
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
6.
J Forensic Sci ; 69(4): 1171-1182, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38798041

ABSTRACT

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.


Subject(s)
Firearms , Forensic Ballistics , Head Injuries, Penetrating , Wounds, Gunshot , Humans , Wounds, Gunshot/pathology , Male , Head Injuries, Penetrating/pathology , Barium/analysis , Lead/analysis , Adult , Middle Aged , Homicide , Skull/injuries , Skull/pathology , Cyclohexanones
7.
Clin Exp Dent Res ; 10(3): e875, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38798121

ABSTRACT

BACKGROUND: The bone regeneration therapy is often used in patients with inadequate bone support for implants, particularly following tooth extractions. Xenografts derived from animal tissues are effective bone reconstructive options that resist resorption and pose a low risk of transmitting disease. Therefore, these implants may be a good option for enhancing and stabilizing maxillary sinuses. The purpose of this study was to compare two xenografts, Bone+B® and InterOss®, for the reconstruction of rabbit calvaria defects. METHODS AND MATERIALS: The study involved seven male New Zealand white rabbits. In the surgical procedure, 21 spots were created on both sides of the midline calvarium by creating three 8-millimeter defects. A control group was used, as well as two treatment groups utilizing Bone+B® Grafts and InterOss® Grafts. After 3 months, the rabbits were euthanized, followed by pathological evaluation. Analysis of these samples focused on bone formation, xenograft remaining material, and inflammation levels, using Adobe Photoshop CS 8.0 and SPSS version 24. RESULTS: With the application of Bone+B® graft, bone formation ranged from 32% to 45%, with a mean of 37.80% (±5.63), and the remaining material ranged from 28% to 37%, with a mean of 32.60% (±3.65). Using InterOss® grafts, bone formation was 61% to 75%, the mean was 65.83% (±4.75), and the remaining material was 9% to 18%, with a mean of 13.17% (±3.06). The bone formation in the control group ranged from 10% to 25%, with a mean of 17.17% (±6.11). InterOss® had lower inflammation levels than other groups, but the difference was not statistically significant (p > .05). CONCLUSION: InterOss® bone powder is the best option for maxillofacial surgery and bone reconstruction. This is due to more bone formation, less remaining material, and a lower inflammation level. Compared to the control group, Bone+B® improves healing and bone quality, thus making it an alternative to InterOss®.


Subject(s)
Bone Regeneration , Bone Substitutes , Bone Transplantation , Heterografts , Skull , Animals , Rabbits , Skull/surgery , Skull/pathology , Male , Bone Transplantation/methods , Bone Substitutes/pharmacology , Osteogenesis
8.
Biomacromolecules ; 25(6): 3784-3794, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38743836

ABSTRACT

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.


Subject(s)
Bone Regeneration , Mesenchymal Stem Cells , Osteogenesis , Polyesters , Tissue Engineering , Tissue Scaffolds , Animals , Tissue Scaffolds/chemistry , Rats , Bone Regeneration/drug effects , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Osteogenesis/physiology , Tissue Engineering/methods , Polyesters/chemistry , Cell Differentiation , Rats, Sprague-Dawley , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cells, Cultured , Cell Proliferation , Skull/injuries , Skull/pathology , Durapatite/chemistry , Durapatite/pharmacology
9.
Mol Biol Rep ; 51(1): 632, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724827

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) play critical roles in the osteogenic differentiation of human bone mesenchymal stem cells (hBMSCs), but the mechanism by which miRNAs indirectly modulate osteogenesis remains unclear. Here, we explored the mechanism by which miRNAs indirectly modulate gene expression through histone demethylases to promote bone regeneration. METHODS AND RESULTS: Bioinformatics analysis was performed on hBMSCs after 7 days of osteogenic induction. The differentially expressed miRNAs were screened, and potential target mRNAs were identified. To determine the bioactivity and stemness of hBMSCs and their potential for bone repair, we performed wound healing, Cell Counting Kit-8 (CCK-8), real-time reverse transcription quantitative polymerase chain reaction (RT‒qPCR), alkaline phosphatase activity, alizarin red S (ARS) staining and radiological and histological analyses on SD rats with calvarial bone defects. Additionally, a dual-luciferase reporter assay was utilized to investigate the interaction between miR-26b-5p and ten-eleven translocation 3 (TET3) in human embryonic kidney 293T cells. The in vitro and in vivo results suggested that miR-26b-5p effectively promoted the migration, proliferation and osteogenic differentiation of hBMSCs, as well as the bone reconstruction of calvarial defects in SD rats. Mechanistically, miR-26b-5p bound to the 3' untranslated region of TET3 mRNA to mediate gene silencing. CONCLUSIONS: MiR-26b-5p downregulated the expression of TET3 to increase the osteogenic differentiation of hBMSCs and bone repair in rat calvarial defects. MiR-26b-5p/TET3 crosstalk might be useful in large-scale critical bone defects.


Subject(s)
Dioxygenases , Mesenchymal Stem Cells , MicroRNAs , Osteogenesis , Animals , Female , Humans , Rats , Bone Regeneration/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Dioxygenases/genetics , Dioxygenases/metabolism , HEK293 Cells , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Osteogenesis/genetics , Rats, Sprague-Dawley , Skull/pathology , Skull/metabolism
10.
Colloids Surf B Biointerfaces ; 239: 113969, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749168

ABSTRACT

In this study, poly (lactic acid)/zinc-doped nano hydroxyapatite (PLA/nano-ZnHA) composite microspheres were prepared and formed into injectable bone paste with sodium alginate (SA) and polyvinyl alcohol (PVA) for bone defect repair. The effect of component of bone paste on injectability and zinc doping content related biological properties were mainly discussed. An injectable bone paste of PLA/nano-ZnHA composite microspheres (CM) was formed in mass ratio of (2.5-25):(0.25-4): (0-2.5):(20-65) of CM, SA, PVA and water with the favorable injectability (average force:4.46±1.72 N). In vitro 5%-10% zinc doping content displayed significantly higher promotion on cell proliferation and osteogenic differentiation than 15%-20% zinc doping content. Furthermore, in vivo the significant promoting effect of 0-5% zinc doping in ZnHA on bone repair was observed. Although 5% zinc doping content did not show a significant enhancement in bone volume/tissue volume (BV/TV), it has the ability to improve the bone mineral density (BMD) in early stage of bone repair compared with the 0% zinc doping content. The PLA/nano-ZnHA composite microsphere injectable paste with convenient surgical operation and well filling ability has the potential to become a competitive tissue repair material.


Subject(s)
Durapatite , Microspheres , Polyesters , Skull , Zinc , Polyesters/chemistry , Durapatite/chemistry , Durapatite/pharmacology , Zinc/chemistry , Zinc/pharmacology , Animals , Skull/drug effects , Skull/pathology , Cell Proliferation/drug effects , Osteogenesis/drug effects , Injections , Cell Differentiation/drug effects
11.
Colloids Surf B Biointerfaces ; 239: 113971, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38759296

ABSTRACT

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.


Subject(s)
Alginates , Catechin , Cell Proliferation , Hydrogels , Skull , Tissue Scaffolds , Zinc , Zinc/chemistry , Zinc/pharmacology , Alginates/chemistry , Alginates/pharmacology , Catechin/chemistry , Catechin/analogs & derivatives , Catechin/pharmacology , Skull/drug effects , Skull/injuries , Skull/pathology , Animals , Mice , Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Scaffolds/chemistry , Cell Proliferation/drug effects , Osteoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/metabolism , Cell Differentiation/drug effects , Osteogenesis/drug effects , Bone Regeneration/drug effects , Cell Adhesion/drug effects
12.
Int J Biol Macromol ; 270(Pt 1): 132361, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38750857

ABSTRACT

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.


Subject(s)
Bone Regeneration , Glass , Osteogenesis , Polyesters , Printing, Three-Dimensional , Rats, Wistar , Skull , Tissue Scaffolds , Animals , Polyesters/chemistry , Tissue Scaffolds/chemistry , Rats , Bone Regeneration/drug effects , Skull/drug effects , Skull/pathology , Skull/injuries , Skull/diagnostic imaging , Osteogenesis/drug effects , Female , Glass/chemistry , Tragacanth/chemistry , X-Ray Microtomography , Tissue Engineering/methods , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
13.
J Transl Med ; 22(1): 437, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720345

ABSTRACT

BACKGROUND: Biological-derived hydroxyapatite is widely used as a bone substitute for addressing bone defects, but its limited osteoconductive properties necessitate further improvement. The osteo-immunomodulatory properties hold crucial promise in maintaining bone homeostasis, and precise modulation of macrophage polarization is essential in this process. Metabolism serves as a guiding force for immunity, and fluoride modification represents a promising strategy for modulating the osteoimmunological environment by regulating immunometabolism. In this context, we synthesized fluorinated porcine hydroxyapatite (FPHA), and has demonstrated its enhanced biological properties and osteogenic capacity. However, it remains unknown whether and how FPHA affects the immune microenvironment of the bone defects. METHODS: FPHA was synthesized and its composition and structural properties were confirmed. Macrophages were cultured with FPHA extract to investigate the effects of FPHA on their polarization and the related osteo-immune microenvironment. Furthermore, total RNA of these macrophages was extracted, and RNA-seq analysis was performed to explore the underlying mechanisms associated with the observed changes in macrophages. The metabolic states were evaluated with a Seahorse analyzer. Additionally, immunohistochemical staining was performed to evaluate the macrophages response after implantation of the novel bone substitutes in critical size calvarial defects in SD rats. RESULTS: The incorporation of fluoride ions in FPHA was validated. FPHA promoted macrophage proliferation and enhanced the expression of M2 markers while suppressing the expression of M1 markers. Additionally, FPHA inhibited the expression of inflammatory factors and upregulated the expression of osteogenic factors, thereby enhancing the osteogenic differentiation capacity of the rBMSCs. RNA-seq analysis suggested that the polarization-regulating function of FPHA may be related to changes in cellular metabolism. Further experiments confirmed that FPHA enhanced mitochondrial function and promoted the metabolic shift of macrophages from glycolysis to oxidative phosphorylation. Moreover, in vivo experiments validated the above results in the calvarial defect model in SD rats. CONCLUSION: In summary, our study reveals that FPHA induces a metabolic shift in macrophages from glycolysis to oxidative phosphorylation. This shift leads to an increased tendency toward M2 polarization in macrophages, consequently creating a favorable osteo-immune microenvironment. These findings provide valuable insights into the impact of incorporating an appropriate concentration of fluoride on immunometabolism and macrophage mitochondrial function, which have important implications for the development of fluoride-modified immunometabolism-based bone regenerative biomaterials and the clinical application of FPHA or other fluoride-containing materials.


Subject(s)
Durapatite , Glycolysis , Macrophages , Oxidative Phosphorylation , Rats, Sprague-Dawley , Animals , Durapatite/chemistry , Macrophages/metabolism , Macrophages/drug effects , Oxidative Phosphorylation/drug effects , Glycolysis/drug effects , Rats , Swine , Cell Proliferation/drug effects , Male , Osteogenesis/drug effects , Skull/pathology , Skull/drug effects , Mice , Cellular Microenvironment/drug effects , RAW 264.7 Cells , Bone and Bones/metabolism , Bone and Bones/drug effects
14.
J Vis Exp ; (206)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38709029

ABSTRACT

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.


Subject(s)
Brain Concussion , Disease Models, Animal , Skull , Animals , Mice , Brain Concussion/pathology , Skull/pathology , Skull/injuries , Skull/surgery , Male , Percussion/methods , Brain Injuries, Traumatic/pathology
15.
Jt Dis Relat Surg ; 35(2): 354-360, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38727115

ABSTRACT

OBJECTIVES: This study aims to compare cranial bone ossification between patients with developmental dysplasia of the hip (DDH) and healthy individuals. PATIENTS AND METHODS: Between September 2021 and April 2022, a total of 60 healthy female individuals (median age: 24.5 months; range, 18 to 36 months) and 56 female DDH patients (median age: 23 months; range, 18 to 35 months) were included. Age, head circumference, weight, height, and patency of the anterior fontanel were measured in groups. Percentiles were classified as very low, low, normal, high and very high. All patients were female and those with abnormal thyroid function test, vitamin D, calcium, phosphate and alkaline phosphatase values were not included in the study. For those diagnosed with DDH, they were included in the group regardless of the type of treatment. RESULTS: No statistically significant difference was found between the groups in terms of age and weight (p>0.05). The very low and very high head circumferences were more frequent, and the normal head circumferences were less frequent in the DDH group (p<0.05). There was no significant difference between groups in terms of fontanel closure (p>0.05). In open fontanels, no significant difference was found in both groups in terms of age (p>0.05). CONCLUSION: Our study results showed no significant difference between the fontanel ossifications of children with and without DDH; however, we found that the ossification of the skull bones of children with DDH was different compared to healthy children.


Subject(s)
Developmental Dysplasia of the Hip , Osteogenesis , Skull , Humans , Female , Child, Preschool , Infant , Developmental Dysplasia of the Hip/surgery , Developmental Dysplasia of the Hip/pathology , Developmental Dysplasia of the Hip/diagnostic imaging , Skull/pathology , Skull/growth & development , Skull/diagnostic imaging , Osteogenesis/physiology , Case-Control Studies
16.
Int J Biol Macromol ; 269(Pt 1): 131914, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703527

ABSTRACT

The healing of critical-sized bone defects is a major challenge in the field of bone tissue engineering. Gelatin-related hydrogels have emerged as a potential solution due to their desirable properties. However, their limited osteogenic, mechanical, and reactive oxygen species (ROS)-scavenging capabilities have hindered their clinical application. To overcome this issue, we developed a biofunctional gelatin-Mxene nanocomposite hydrogel. Firstly, we prepared two-dimensional (2D) Ti3C2 MXene nanosheets using a layer delamination method. Secondly, these nanosheets were incorporated into a transglutaminase (TG) enzyme-containing gallic acid-imbedded gelatin (GGA) pre-gel solution to create an injectable GGA-MXene (GM) nanocomposite hydrogel. The GM hydrogels exhibited superior compressive strength (44-75.6 kPa) and modulus (24-44.5 kPa) compared to the GGA hydrogels. Additionally, the GM hydrogel demonstrated the ability to scavenge reactive oxygen species (OH- and DPPH radicals), protecting MC3T3-E1 cells from oxidative stress. GM hydrogels were non-toxic to MC3T3-E1 cells, increased alkaline phosphatase secretion, calcium nodule formation, and upregulated osteogenic gene expressions (ALP, OCN, and RUNX2). The GM400 hydrogel was implanted in critical-sized calvarial defects in rats. Remarkably, it exhibited significant potential for promoting new bone formation. These findings indicated that GM hydrogel could be a viable candidate for future clinical applications in the treatment of critical-sized bone defects.


Subject(s)
Gelatin , Hydrogels , Nanocomposites , Osteogenesis , Reactive Oxygen Species , Skull , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Gelatin/chemistry , Nanocomposites/chemistry , Osteogenesis/drug effects , Reactive Oxygen Species/metabolism , Skull/drug effects , Skull/pathology , Mice , Rats , Bone Regeneration/drug effects , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Titanium/chemistry , Cell Line , Tissue Engineering/methods
17.
Orphanet J Rare Dis ; 19(1): 204, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762603

ABSTRACT

BACKGROUND: Trigonocephaly occurs due to the premature fusion of the metopic suture, leading to a triangular forehead and hypotelorism. This condition often requires surgical correction for morphological and functional indications. Metopic ridges also originate from premature metopic closure but are only associated with mid-frontal bulging; their surgical correction is rarely required. Differential diagnosis between these two conditions can be challenging, especially in minor trigonocephaly. METHODS: Two hundred seven scans of patients with trigonocephaly (90), metopic rigdes (27), and controls (90) were collected. Geometric morphometrics were used to quantify skull and orbital morphology as well as the interfrontal angle and the cephalic index. An innovative method was developed to automatically compute the frontal curvature along the metopic suture. Different machine-learning algorithms were tested to assess the predictive power of morphological data in terms of classification. RESULTS: We showed that control patients, trigonocephaly and metopic rigdes have distinctive skull and orbital shapes. The 3D frontal curvature enabled a clear discrimination between groups (sensitivity and specificity > 92%). Furthermore, we reached an accuracy of 100% in group discrimination when combining 6 univariate measures. CONCLUSION: Two diagnostic tools were proposed and demonstrated to be successful in assisting differential diagnosis for patients with trigonocephaly or metopic ridges. Further clinical assessments are required to validate the practical clinical relevance of these tools.


Subject(s)
Craniosynostoses , Humans , Craniosynostoses/diagnostic imaging , Craniosynostoses/pathology , Craniosynostoses/diagnosis , Female , Male , Infant , Imaging, Three-Dimensional/methods , Skull/diagnostic imaging , Skull/pathology
18.
J Craniofac Surg ; 35(4): e396-e399, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38738899

ABSTRACT

This study combines clinical and anthropological analyses to investigate the complex cranial pathology of a South African individual from the 19th century. The cranium was examined macroscopically and radiographically. Conducting a standard differential diagnosis was challenging given the complexity and uncommon nature of the pathology and required drawing on relatively sparse paleopathological and clinical case reports. Multiple conditions were identified including biparietal thinning, basilar invagination, platybasia, and complicated chronic frontal sinusitis, where the intracranial extension of sinus infection may likely have contributed to the individual's death. The authors urge for awareness of these uncommon conditions, as their presence can easily be overlooked or confound skeletal assessments. This clinical study contributes to the authors' understanding of uncommon and poorly described paleopathological diseases and will help to better facilitate their diagnosis in future research. It represents one of the first studies describing such an unusual cooccurrence of uncommon pathologies in an archeological individual.


Subject(s)
Skull , Humans , South Africa , History, 19th Century , Skull/diagnostic imaging , Skull/pathology , Male , Paleopathology , Tomography, X-Ray Computed , Diagnosis, Differential
19.
Carbohydr Polym ; 336: 122115, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38670750

ABSTRACT

To alleviate skull defects and enhance the biological activity of taxifolin, this study utilized the thin-film dispersion method to prepare paclitaxel liposomes (TL). Thiolated chitosan (CSSH)-modified TL (CTL) was synthesized through charge interactions. Injectable hydrogels (BLG) were then prepared as hydrogel scaffolds loaded with TAX (TG), TL (TLG), and CTL (CTLG) using a Schiff base reaction involving oxidized dextran and carboxymethyl chitosan. The study investigated the bone reparative properties of CTLG through molecular docking, western blot techniques, and transcriptome analysis. The particle sizes of CTL were measured at 248.90 ± 14.03 nm, respectively, with zeta potentials of +36.68 ± 5.43 mV, respectively. CTLG showed excellent antioxidant capacity in vitro. It also has a good inhibitory effect on Escherichia coli and Staphylococcus aureus, with inhibition rates of 93.88 ± 1.59 % and 88.56 ± 2.83 % respectively. The results of 5-ethynyl-2 '-deoxyuridine staining, alkaline phosphatase staining and alizarin red staining showed that CTLG also had the potential to promote the proliferation and differentiation of mouse embryonic osteoblasts (MC3T3-E1). The study revealed that CTLG enhances the expression of osteogenic proteins by regulating the Wnt signaling pathway, shedding light on the potential application of TAX and bone regeneration mechanisms.


Subject(s)
Cell Proliferation , Chitosan , Hydrogels , Liposomes , Osteoblasts , Quercetin , Quercetin/analogs & derivatives , Skull , Wnt Signaling Pathway , Animals , Chitosan/analogs & derivatives , Chitosan/chemistry , Chitosan/pharmacology , Quercetin/pharmacology , Quercetin/chemistry , Liposomes/chemistry , Wnt Signaling Pathway/drug effects , Osteoblasts/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Cell Proliferation/drug effects , Mice , Skull/drug effects , Skull/pathology , Skull/metabolism , Rats , Bone Regeneration/drug effects , Rats, Sprague-Dawley , Osteogenesis/drug effects , Staphylococcus aureus/drug effects , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Cell Differentiation/drug effects , Escherichia coli/drug effects , Male , Molecular Docking Simulation
20.
Biomed Pharmacother ; 175: 116606, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670048

ABSTRACT

Stress-induced premature senescent (SIPS) cells induced by various stresses deteriorate cell functions. Dasatinib and quercetin senolytics (DQ) can alleviate several diseases by eliminating senescent cells. α-tricalcium phosphate (α-TCP) is a widely used therapeutic approach for bone restoration but induces bone formation for a comparatively long time. Furthermore, bone infection exacerbates the detrimental prognosis of bone formation during material implant surgery due to oral cavity bacteria and unintentional contamination. It is essential to mitigate the inhibitory effects on bone formation during surgical procedures. Little is known that DQ improves bone formation in Lipopolysaccharide (LPS)-contaminated implants and its intrinsic mechanisms in the study of maxillofacial bone defects. This study aims to investigate whether the administration of DQ ameliorates the impairments on bone repair inflammation and contamination by eliminating SIPS cells. α-TCP and LPS-contaminated α-TCP were implanted into Sprague-Dawley rat calvaria bone defects. Simultaneously, bone formation in the bone defects was investigated with or without the oral administration of DQ. Micro-computed tomography and hematoxylin-eosin staining showed that senolytics significantly enhanced bone formation at the defect site. Histology and immunofluorescence staining revealed that the levels of p21- and p16-positive senescent cells, inflammation, macrophages, reactive oxygen species, and tartrate-resistant acid phosphatase-positive cells declined after administering DQ. DQ could partially alleviate the production of senescent markers and senescence-associated secretory phenotypes in vitro. This study indicates that LPS-contaminated α-TCP-based biomaterials can induce cellular senescence and hamper bone regeneration. Senolytics have significant therapeutic potential in reducing the adverse osteogenic effects of biomaterial-related infections and improving bone formation capacity.


Subject(s)
Bone Regeneration , Cellular Senescence , Inflammation , Osteogenesis , Rats, Sprague-Dawley , Senotherapeutics , Signal Transduction , Animals , Bone Regeneration/drug effects , Cellular Senescence/drug effects , Senotherapeutics/pharmacology , Signal Transduction/drug effects , Inflammation/drug therapy , Inflammation/pathology , Osteogenesis/drug effects , Rats , Male , Quercetin/pharmacology , Dasatinib/pharmacology , Lipopolysaccharides , Skull/drug effects , Skull/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...