Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.338
Filtrar
1.
Bioact Mater ; 43: 98-113, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39381328

RESUMO

More than 500,000 bone grafting procedures are performed annually in the USA. Considering the significant limitations of available bone grafts, we previously invented a phase-separation technology to generate nanofibrous poly(l-lactic acid) (PLLA) scaffolds that mimic the bone matrix collagen in nanofiber geometry and enhance bone regeneration. Here we report the development of nanofibrous scaffolds with covalently attached synthetic peptides that mimic native collagen peptides to activate the two main collagen receptors in bone cells, discoidin domain receptor 2 (DDR2) and ß1 integrins. We synthesized a PLLA-based graft-copolymer to enable covalent peptide conjugation via a click reaction. Using PLLA and the graft-copolymer, we developed 3D scaffolds with interconnected pores and peptides-containing nanofibers to activate DDR2 and ß1 integrins of osteogenic cells. The degradation rate and mechanical properties of the scaffolds are tunable. The peptides-decorated nanofibrous scaffolds demonstrated 7.8 times more mineralized bone regeneration over the control scaffolds without the peptides in a critical-sized bone defect regeneration model after 8 weeks of implantation, showing a synergistic effect of the two peptides. This study demonstrates the power of scaffolds to mimic ECM at both nanometer and molecular levels, activating cell surface receptors to liberate the innate regenerative potential of host stem/progenitor cells.

2.
Front Pharmacol ; 15: 1462368, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39359247

RESUMO

As a novel class of smart biomaterials with promising potentials, metal-organic frameworks (MOFs) are widely utilized in the field of biomedicine. Current researches indicate that the therapeutic strategies for osteoarthritis (OA) are highly limited to achieving symptom improvement and reducing both pain and inflammation. Together, the introduction of MOFs into the treatment of OA holds the potential to offer significant benefits. This is because MOFs not only have intrinsic biological activities, but also act as carriers to facilitate controlled drug delivery and prolong the duration in the management of OA. This paper presents a review of the recent studies that have explored the potential usage of MOFs as drugs or carriers in the treatment of OA, which also examines the progress of MOFs in tissue engineering for the treatment of OA. These studies are anticipated to not only enhance the comprehension of MOFs but also provide strong evidence in favor of their utilization in the treatment of OA.

3.
Int J Biol Macromol ; 281(Pt 2): 136384, 2024 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-39383920

RESUMO

Nowadays, bone injuries and disorders have increased all over the world and can reduce the quality of human life. Bone tissue engineering repair approaches require new biomaterials and methods to construct scaffolds with the required structural properties as well as improved performance. As potential therapeutic strategies in bone tissue engineering, 3D printed scaffolds have been developed. Polycaprolactone/Ceramic composites have attracted considerable attention due to their cytocompatibility, biodegradability, and physical properties. In this study, a 3D printing process was used to create polycaprolactone (PCL)-Gelatin (GEL) scaffolds containing varying concentrations of Bioglass (BG) and Nano Montmorillonite (MMT). This mixture was then loaded into a 3D printer, and the scaffolds were printed layer by layer. After constructing the scaffolds, they were then examined for their physical, chemical, and biological characteristics. Surface appearance was analyzed with a scanning electron microscope (SEM), which revealed that NC increased the diameter of pores from 465 to 480 µm. The elements in the scaffolds were evaluated by EDX analysis, and a uniform dispersion of nano montmorillonite particles was observed. The compressive strength reached 76.43 MPa for PCL/G/35 %MMT/15 %BG scaffold. Also, the rate of water absorption, biodegradability and bioactivity of PCL-GEL scaffolds increased significantly in the presence of NC. According to the MTT cell test results, adding BG and NC increased cell proliferation, adhesion and cell viability to 127.7 %. These findings indicated that the 3D printed PCL/G/35 %MMT/15 %BG scaffold has promising strategies for bone repair applications. Also, polynomial curve fitting shows that scaffold degradability after soaking in PBS can be predicted using the initial weight and soaking time. Adding more variables and data could improve prediction accuracy, reducing the need for experiments and conserving resources.

4.
Macromol Biosci ; : e2400279, 2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39388643

RESUMO

The combination of gelatin and hydroxyapatite (HA) has emerged as a promising strategy in dental tissue engineering due to its favorable biocompatibility, mechanical properties, and ability to support cellular activities essential for tissue regeneration, rendering them ideal components for hard tissue applications. Besides, precise control over interconnecting porosity is of paramount importance for tissue engineering materials. Conventional methods for creating porous scaffolds frequently encounter difficulties in regulating pore size distribution. This study demonstrates the fabrication of gelatin-nano HA scaffolds with uniform porosity using a T-type junction microfluidic device in a single-step process. Significant improvements in control over the pore size distribution are achieved by regulating the flow parameters, resulting in effective and time-efficient manufacturing comparable in quality to the innovative 3D bioprinting techniques. The overall porosity of the scaffolds exceeded 60%, with a remarkably narrow size distribution. The incorporation of nano-HAinto 3D porous gelatin scaffolds successfully induced osteogenic differentiation in stem cells at both the protein and gene levels, as evidenced by the significant increase in osteocalcin (OCN), an important marker of osteogenic differentiation. The OCN levels are 26 and 43 times higher for gelatin and gelatin-HA scaffolds, respectively, compared to the control group.

5.
ACS Appl Bio Mater ; 2024 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-39382202

RESUMO

For better bone regeneration, precise control over the architecture of the scaffolds is necessary. Because the shape of the pore may affect the bone regeneration, therefore, additive manufacturing has been used in this study to fabricate magnetic bioactive glass (MBG) scaffolds with three different architectures, namely, grid, gyroid, and Schwarz D surface with 15 × 15 × 15 mm3 dimensions and 70% porosity. These scaffolds have been fabricated using an in-house-developed material-extrusion-based additive manufacturing system. The composition of bioactive glass was selected as 45% SiO2, 20% Na2O, 23% CaO, 6% P2O5, 2.5% B2O3, 1% ZnO, 2% MgO, and 0.5% CaF2 (wt %), and additionally 0.4 wt % of iron carbide nanoparticles were incorporated. Afterward, MBG powder was mixed with a 25% (w/v) Pluronic F-127 solution to prepare a slurry for fabricating scaffolds at 23% relative humidity. The morphological characterization using microcomputed tomography revealed the appropriate pore size distribution and interconnectivity of the scaffolds. The compressive strengths of the fabricated grid, gyroid, and Schwarz D scaffolds were found to be 14.01 ± 1.01, 10.78 ± 1.5, and 12.57 ± 1.2 MPa, respectively. The in vitro study was done by immersing the MBG scaffolds in simulated body fluid for 1, 3, 7, and 14 days. Darcy's law, which describes the flow through porous media, was used to evaluate the permeability of the scaffolds. Furthermore, an anticancer drug (Mitomycin C) was loaded onto these scaffolds, wherein these scaffolds depicted good release behavior. Overall, gyroid-structured scaffolds were found to be the most suitable among the three scaffolds considered in this study for bone tissue engineering and drug-delivery applications.

6.
Int J Biol Macromol ; 281(Pt 2): 136340, 2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-39374728

RESUMO

Novel biomaterials are necessary to fabricate biomimetic scaffolds for bone tissue engineering. In the present experiment, we aimed to fabricate and evaluate the osteogenic properties of nanohydroxyapatite/chitosan/decellularized placenta (nHA.Cs.dPL) composite scaffolds. The human placenta was decellularized (dPL), characterized, and digested in pepsin to form the hydrogel. nHA.Cs.dPL scaffolds were fabricated using salt leaching/freeze drying and evaluated for their morphology, chemical composition, swelling, porosity, degradation, mechanical strength, and biocompatibility. Saos-2 cells were seeded on scaffolds, and their osteogenic properties were investigated by evaluating alkaline phosphatase (ALP), osteocalcin (OCN), collagen type 1 (COL I) expression, and calcium deposition under osteogenic differentiation. The dPL was prepared with minimized DNA content and a well-preserved porous structure. Scaffolds were highly porous with interconnected pores and exhibited appropriate swelling and degradation rates supporting saos-2 cell attachment and proliferation. dPL improved scaffold physicochemical features and increased cell proliferation, ALP, OCN, COL I expression, and calcium deposition under osteogenic differentiation induction. nHA.Cs.dPL composite scaffolds provide a 3D microenvironment with superior physicochemical features that support saos-2 cell adhesion, proliferation, and osteogenic differentiation.

7.
Int J Biol Macromol ; 281(Pt 1): 136160, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39357695

RESUMO

Recently, the fabrication of personalized scaffolds with high accuracy has been developed through 3D printing technology. In the current study, polylactic acid/polyethylene glycol (PLA/PEG) composite scaffolds with varied weight percentages (0, 5, 10, 20 and 30 %) of bredigite nanoparticles (B) were fabricated using the 3D printing and then characterized through scanning electron microscopy and Fourier transform infra-red spectroscopy. The addition of B nanoparticles up to 20 wt% to PLA/PEG scaffold increased the compressive strength (from 7.59 to 13.84 MPa) and elastic modulus (from 142.42 to 268.33 MPa). The apatite formation ability as well as inorganic ion release in simulated body fluid were investigated for 28 days. The MG-63 cells viability and adhesion were enhanced by increasing the amount of B in the PLA/PEG scaffold and the osteogenic differentiation of the rat bone marrow mesenchymal stem cells was confirmed by alkaline phosphatase activity test and alizarin red staining. According to chorioallantoic membrane assay, the highest angiogenesis occurred around the PLA/PEG/B30 scaffold. In vivo experiments on a rat calvarial defect model demonstrated an almost complete recovery in the PLA/PEG/B30 group within 8 weeks. Based on the results, the PLA/PEG/B30 composite scaffold is proposed as an optimal scaffold to repair bone defects.

8.
Int J Biol Macromol ; : 136530, 2024 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-39406323

RESUMO

Chitosan, a sustainable and highly abundant animal-derived biopolymer, possesses versatile properties, such as solubility, film-forming ability, viscosity, ion binding, and antimicrobial qualities, which are suitable for biomedical applications. Due to its charged nature, chitosan is a lucrative biopolymer for scaffold fabrication, especially for bone-tissue engineering applications, using the electrospinning method, which is an industrially suitable, scalable, and swift method for fabricating porous nanocomposite structures. Despite a lot of research being conducted on chitosan-based electrospun materials for bone tissue engineering, the research on this topic has not been thoroughly reviewed. This review article aims to fill this knowledge gap and provides an in-depth discussion of the research on this topic. To start with, a brief overview of bone tissue engineering has been provided, followed by the properties of chitosan, which make it an important biopolymer for this application. Also, the important factors that must be considered while electrospinning chitosan, especially considering its application in bone tissue engineering, have been debated. Further, the type of chitosan-based electrospun material has been discussed along with the recent advancements in this research area. Finally, a brief perspective on the future of this technology has been provided.

10.
Polymers (Basel) ; 16(19)2024 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-39408507

RESUMO

Bone repair and regeneration require physiological cues, including mechanical, electrical, and biochemical activity. Many biomaterials have been investigated as bioactive scaffolds with excellent electrical properties. Amongst biomaterials, piezoelectric materials (PMs) are gaining attention in biomedicine, power harvesting, biomedical devices, and structural health monitoring. PMs have unique properties, such as the ability to affect physiological movements and deliver electrical stimuli to damaged bone or cells without an external power source. The crucial bone property is its piezoelectricity. Bones can generate electrical charges and potential in response to mechanical stimuli, as they influence bone growth and regeneration. Piezoelectric materials respond to human microenvironment stimuli and are an important factor in bone regeneration and repair. This manuscript is an overview of the fundamentals of the materials generating the piezoelectric effect and their influence on bone repair and regeneration. This paper focuses on the state of the art of piezoelectric materials, such as polymers, ceramics, and composites, and their application in bone tissue engineering. We present important information from the point of view of bone tissue engineering. We highlight promising upcoming approaches and new generations of piezoelectric materials.

11.
J Funct Biomater ; 15(9)2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39330217

RESUMO

The rising demand for effective bone regeneration has underscored the limitations of traditional methods like autografts and allografts, including donor site morbidity and insufficient biological signaling. This review examines nanoparticles (NPs) in tissue engineering (TE) to address these challenges, evaluating polymers, metals, ceramics, and composites for their potential to enhance osteogenesis and angiogenesis by mimicking the extracellular matrix (ECM) nanostructure. The methods involved synthesizing and characterizing nanoparticle-based scaffoldsand integrating hydroxyapatite (HAp) with polymers to enhance mechanical properties and osteogenic potential. The results showed that these NPs significantly promote cell growth, differentiation, and bone formation, with carbon-based NPs like graphene and carbon nanotubes showing promise. NPs offer versatile, biocompatible, and customizable scaffolds that enhance drug delivery and support bone repair. Despite promising results, challenges with cytotoxicity, biodistribution, and immune responses remain. Addressing these issues through surface modifications and biocompatible molecules can improve the biocompatibility and efficacy of nanomaterials. Future research should focus on long-term in vivo studies to assess the safety and efficacy of NP-based scaffolds and explore synergistic effects with other bioactive molecules or growth factors. This review underscores the transformative potential of NPs in advancing BTE and calls for further research to optimize these technologies for clinical applications.

12.
J Funct Biomater ; 15(9)2024 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-39330219

RESUMO

Bone tissue engineering has seen significant advancements with innovative scaffold fabrication techniques such as 3D printing. This review focuses on enhancing polycaprolactone (PCL) scaffold properties through structural modifications, including surface treatments, pore architecture adjustments, and the incorporation of biomaterials like hydroxyapatite (HA). These modifications aim to improve scaffold conformation, cellular behavior, and mechanical performance, with particular emphasis on the role of mesenchymal stem cells (MSCs) in bone regeneration. The review also explores the potential of integrating nanomaterials and graphene oxide (GO) to further enhance the mechanical and biological properties of PCL scaffolds. Future directions involve optimizing scaffold structures and compositions for improved bone tissue regeneration outcomes.

13.
Nanomaterials (Basel) ; 14(18)2024 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-39330641

RESUMO

Percutaneous implants osseointegrated into the residuum of a person with limb amputation need to provide mechanical stability and protection against infections. Although significant progress has been made in the biointegration of percutaneous implants, the problem of forming a reliable natural barrier at the level of the surface of the implant and the skin and bone tissues remains unresolved. The use of a microporous implant structure incorporated into the Skin and Bone Integrated Pylon (SBIP) should address the issue by allowing soft and bone tissues to grow directly into the implant structure itself, which, in turn, should form a reliable barrier to infections and support strong osseointegration. To evaluate biological interactions between dermal fibroblasts and MC3T3-E1 osteoblasts in vitro, small titanium discs (with varying pore sizes and volume fractions to achieve deep porosity) were fabricated via 3D printing and sintering. The cell viability MTT assay demonstrated low cytotoxicity for cells co-cultured in the pores of the 3D-printed and sintered Ti samples during the 14-day follow-up period. A subsequent Quantitative Real-Time Polymerase Chain Reaction (RT-PCR) analysis of the relative gene expression of biomarkers that are associated with cell adhesion (α2, α5, αV, and ß1 integrins) and extracellular matrix components (fibronectin, vitronectin, type I collagen) demonstrated that micropore sizes ranging from 200 to 500 µm of the 3D printed and sintered Ti discs were favorable for dermal fibroblast adhesion. For example, for representative 3D-printed Ti sample S6 at 72 h the values were 4.71 ± 0.08 (α2 integrin), 4.96 ± 0.08 (α5 integrin), 4.71 ± 0.08 (αV integrin), and 1.87 ± 0.12 (ß1 integrin). In contrast, Ti discs with pore sizes ranging from 400 to 800 µm demonstrated the best results (in terms of marker expression related to osteogenic differentiation, including osteopontin, osteonectin, osteocalcin, TGF-ß1, and SMAD4) for MC3T3-E1 cells. For example, for the representative 3D sample S4 on day 14, the marker levels were 11.19 ± 0.77 (osteopontin), 7.15 ± 0.29 (osteonectin), and 6.08 ± 0.12 (osteocalcin), while for sintered samples the levels of markers constituted 5.85 ± 0.4 (osteopontin), 4.45 ± 0.36 (osteonectin), and 4.46 ± 0.3 (osteocalcin). In conclusion, the data obtained show the high biointegrative properties of porous titanium structures, while the ability to implement several pore options in one structure using 3D printing makes it possible to create personalized implants for the best one-time integration with both skin and bone tissues.

14.
Front Biosci (Landmark Ed) ; 29(9): 326, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39344336

RESUMO

BACKGROUND: The development of biomaterials capable of accelerating bone wound repair is a critical focus in bone tissue engineering. This study aims to evaluate the osteointegration and bone regeneration potential of a novel multilayer gelatin-supported Bone Morphogenetic Protein 9 (BMP-9) coated nano-calcium-deficient hydroxyapatite/poly-amino acid (n-CDHA/PAA) composite biomaterials, focusing on the material-bone interface, and putting forward a new direction for the research on the interface between the coating material and bone. METHODS: The BMP-9 recombinant adenovirus (Adenovirus (Ad)-BMP-9/Bone Marrow Mesenchymal Stem Cells (BMSc)) was produced by transfecting BMSc and supported using gelatin (Ad-BMP-9/BMSc/Gelatin (GT). Multilayer Ad-BMP-9/BMSc/GT coated nano-calcium deficient hydroxyapatite/polyamino acid (n-CDHA/PAA) composite biomaterials were then prepared and co-cultured with MG63 cells for 10 days, with biocompatibility assessed through microscopy, Cell Counting Kit-8 (CCK-8), and alkaline phosphatase (ALP) assays. Subsequently, multilayer Ad-BMP-9/BMSc/GT coated n-CDHA/PAA composite biomaterial screws were fabricated, and the adhesion of the coating to the substrate was observed using scanning electron microscopy (SEM). In vivo studies were conducted using a New Zealand White rabbit intercondylar femoral fracture model. The experimental group was fixed with screws featuring multilayer Ad-BMP-9/BMSc/GT coatings, while the control groups used medical metal screws and n-CDHA/PAA composite biomaterial screws. Fracture healing was monitored at 1, 4, 12, and 24 weeks, respectively, using X-ray observation, Micro-CT imaging, and SEM. Integration at the material-bone interface and the condition of neo-tissue were assessed through these imaging techniques. RESULTS: The Ad-BMP-9/GT coating significantly enhanced MG63 cell adhesion, proliferation, and differentiation, while increasing BMP-9 expression in vitro. In vivo studies using a rabbit femoral fracture model confirmed the biocompatibility and osteointegration potential of the multilayer Ad-BMP-9/BMSc/GT coated n-CDHA/PAA composite biomaterial screws. Compared to control groups (medical metal screws and n-CDHA/PAA composite biomaterial screws), this material demonstrated faster fracture healing, stronger osteointegration, and facilitated new bone tissue formation with increased calcium deposition at the material-bone interface. CONCLUSION: The multilayer GT-supported BMP-9 coated n-CDHA/PAA composite biomaterials have demonstrated favorable osteogenic cell interface performance, both in vitro and in vivo. This study provides a foundation for developing innovative bone repair materials, holding promise for significant advancements in clinical applications.


Assuntos
Materiais Revestidos Biocompatíveis , Durapatita , Gelatina , Fator 2 de Diferenciação de Crescimento , Células-Tronco Mesenquimais , Osseointegração , Osteogênese , Animais , Fator 2 de Diferenciação de Crescimento/metabolismo , Gelatina/química , Coelhos , Osseointegração/efeitos dos fármacos , Durapatita/química , Durapatita/farmacologia , Humanos , Materiais Revestidos Biocompatíveis/química , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Regeneração Óssea/efeitos dos fármacos , Fraturas do Fêmur/cirurgia , Engenharia Tecidual/métodos , Materiais Biocompatíveis/química
15.
Int J Biol Macromol ; 279(Pt 4): 135224, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39218179

RESUMO

Polyphenol-functionalized biomaterials are significant in the field of bone tissue engineering (BTE) due to their antioxidant, anti-inflammatory, and osteoinductive properties. In this study, a gelatin (Gel)-based scaffold was functionalized with phloridzin (Ph), the primary polyphenol in apple by-products, to investigate its influence on physicochemical and morphological, properties of the scaffold for BTE application. A preliminary assessment of the biological properties of the functionalized scaffold was also undertaken. The Ph-functionalized scaffold (Gel/Ph) exhibited a porous structure with high porosity (71.3 ± 0.3 %), a pore size of 206.5 ± 1.7 µm, and a radical scavenging activity exceeding 70 %. This scaffold with Young's modulus of 10.8 MPa was determined to support cell proliferation and exhibited cytocompatibility with mesenchymal stem cells (MSCs). Incorporating hydroxyapatite nanoparticle (HA) in the Gel/Ph scaffold stimulated the osteogenic differentiation of key osteogenic genes, including Runx2, ALPL, COL1A1, and OSX ultimately promoting mineralization. This research highlights the promising potential of utilizing polyphenolic compounds derived from fruit waste to functionalize scaffolds for BTE applications.


Assuntos
Diferenciação Celular , Gelatina , Células-Tronco Mesenquimais , Osteogênese , Florizina , Engenharia Tecidual , Alicerces Teciduais , Gelatina/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Florizina/farmacologia , Florizina/química , Osteogênese/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Diferenciação Celular/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/citologia , Proliferação de Células/efeitos dos fármacos , Porosidade , Animais , Durapatita/química , Durapatita/farmacologia , Humanos , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia
16.
Int J Biol Macromol ; 279(Pt 4): 135571, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39276883

RESUMO

To develop a biological bone tissue scaffold with uniform pore size and good cell adhesion was both challenging and imperative. We prepared modified cellulose nanocrystals (CNCs) dispersants (K-PCNCs) by ultrasound-assisted alkylation modification. Subsequently, nano-hydroxyapatite (HC-K) was synthesized using K-PCNCs as a dispersant and composited with polyvinyl alcohol (PVA) to prepare the scaffold using the ice template method. The results showed that the water contact angle and degree of substitution (135°, 1.53) of the K-PCNCs were highest when the ultrasound power was 450 W and the time was 2 h. The dispersion of K-PCNCs prepared under this condition was optimal. SEM showed that the pore distribution of the composite scaffolds was more homogeneous than the PVA scaffold. The porosity, equilibrium swelling rate, and mechanical properties of the composite scaffolds increased and then decreased with the increase of HC-K content, and reached the maximum values (56.1 %, 807.7 %, and 0.085 ± 0.004 MPa) at 9 % (w/w) of HC-K content. Cell experiments confirmed scaffold has good cytocompatibility and mineralization capacity. The ALP activity reached 1.71 ± 0.25 (ALP activity/mg protein). In conclusion, the scaffolds we developed have good biocompatibility and mechanical properties and have great potential in promoting bone defect repair.


Assuntos
Diferenciação Celular , Proliferação de Células , Celulose , Durapatita , Nanopartículas , Osteoblastos , Álcool de Polivinil , Engenharia Tecidual , Alicerces Teciduais , Alicerces Teciduais/química , Celulose/química , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Engenharia Tecidual/métodos , Durapatita/química , Álcool de Polivinil/química , Proliferação de Células/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Nanopartículas/química , Porosidade , Animais , Osso e Ossos/citologia , Ondas Ultrassônicas , Camundongos , Linhagem Celular , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Humanos
17.
ACS Biomater Sci Eng ; 10(10): 6451-6464, 2024 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-39269225

RESUMO

The management of extensive bone loss remains a clinical challenge. Numerous studies are underway to develop a combination of biomaterials, biomolecules, and stem cells to address this challenge. In particular, the systemic administration of antibodies against sclerostin, a regulator of bone formation, was recently shown to enhance the bone repair efficiency of dense collagen hydrogels (DCHs) hosting murine dental pulp stem cells (mDPSCs). The aim of the present study was to assess whether these antibodies, encapsulated and released from DCHs, could promote craniofacial bone repair by the local inhibition of sclerostin. In vitro studies showed that antibody loading modified neither the hydrogel structure nor the viability of seeded mDPSCs. When implanted in a mouse calvaria critical-size bone defect, antibody-loaded DCHs showed repair capabilities similar to those of acellular unloaded DCHs combined with antibody injections. Importantly, the addition of mDPSCs provided no further benefit. Altogether, the local delivery of antisclerostin antibodies from acellular dense collagen scaffolds is highly effective for bone repair. The drastic reduction in the required amount of antibody compared to systemic injection should reduce the cost of the procedure, making the strategy proposed here a promising therapeutic approach for large bone defect repair.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Anticorpos , Colágeno , Hidrogéis , Animais , Hidrogéis/química , Hidrogéis/farmacologia , Camundongos , Colágeno/química , Anticorpos/farmacologia , Anticorpos/imunologia , Anticorpos/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Crânio/efeitos dos fármacos , Crânio/patologia , Regeneração Óssea/efeitos dos fármacos , Células-Tronco , Alicerces Teciduais/química , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia
18.
Immun Inflamm Dis ; 12(9): e70011, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39264247

RESUMO

BACKGROUND: Bone regeneration is a well-regulated dynamic process, of which the prominent role of the immune system on bone homeostasis is more and more revealed by recent research. Before fully activation of the bone remodeling cells, the immune system needs to clean up the microenvironment in facilitating the bone repair initiation. Furthermore, this microenvironment must be maintained properly by various mechanisms over the entire bone regeneration process. OBJECTIVE: This review aims to summarize the role of the T-helper 17/Regulatory T cell (Th17/Treg) balance in bone cell remodeling and discuss the relevant progress in bone tissue engineering. RESULTS: The role of the immune response in the early stages of bone regeneration is crucial, especially the impact of the Th17/Treg balance on osteoclasts, mesenchymal stem cells (MSCs), and osteoblasts activity. By virtue of these knowledge advancements, innovative approaches in bone tissue engineering, such as nano-structures, hydrogel, and exosomes, are designed to influence the Th17/Treg balance and thereby augment bone repair and regeneration. CONCLUSION: Targeting the Th17/Treg balance is a promising innovative strategy for developing new treatments to enhance bone regeneration, thus offering potential breakthroughs in bone injury clinics.


Assuntos
Regeneração Óssea , Osso e Ossos , Linfócitos T Reguladores , Células Th17 , Engenharia Tecidual , Humanos , Linfócitos T Reguladores/imunologia , Engenharia Tecidual/métodos , Regeneração Óssea/imunologia , Animais , Células Th17/imunologia , Osso e Ossos/imunologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Remodelação Óssea/imunologia , Osteoblastos/imunologia , Osteoclastos/imunologia , Osteoclastos/metabolismo
19.
ACS Biomater Sci Eng ; 10(10): 6425-6440, 2024 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-39226111

RESUMO

We fabricated three-dimensional (3D)-printed polycaprolactone (PCL) and PCL/graphene oxide (GO) (PGO) scaffolds for bone tissue engineering. An anti-inflammatory and pro-osteogenesis drug dexamethasone (DEX) was adsorbed onto GO and a 3D-printed PGO/DEX (PGOD) scaffold successfully improved drug delivery with a sustained release of DEX from the scaffold up to 1 month. The physicochemical properties of the PCL, PGO, and PGOD scaffolds were characterized by various analytical techniques. The biological response of these scaffolds was studied for adherence, proliferation, and osteogenic differentiation of seeded rabbit adipose-derived stem cells (ASCs) from DNA assays, alkaline phosphatase (ALP) production, calcium quantification, osteogenic gene expression, and immunofluorescence staining of osteogenic marker proteins. The PGOD scaffold was demonstrated to be the best scaffold for maintaining cell viability, cell proliferation, and osteogenic differentiation of ASCs in vitro. In vivo biocompatibility of PGOD was confirmed from subcutaneous implantation in nude mice where ASC-seeded PGOD can form ectopic bones, demonstrated by microcomputed tomography (micro-CT) analysis and immunofluorescence staining. Furthermore, implantation of PGOD/ASCs constructs into critical-sized cranial bone defects in rabbits form tissue-engineered bones at the defect site, observed using micro-CT and histological analysis.


Assuntos
Dexametasona , Grafite , Osteogênese , Poliésteres , Impressão Tridimensional , Células-Tronco , Engenharia Tecidual , Alicerces Teciduais , Animais , Alicerces Teciduais/química , Engenharia Tecidual/métodos , Coelhos , Dexametasona/farmacologia , Grafite/química , Poliésteres/química , Osteogênese/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Tecido Adiposo/citologia , Diferenciação Celular/efeitos dos fármacos , Camundongos Nus , Osso e Ossos/metabolismo , Camundongos , Proliferação de Células/efeitos dos fármacos
20.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 38(9): 1123-1129, 2024 Sep 15.
Artigo em Chinês | MEDLINE | ID: mdl-39300889

RESUMO

Objective: To develop a biodegradable implantable bone material with compatible mechanics with the bone tissue, providing a new biomaterial for clinical bone repair and regeneration. Methods: Silk reinforced polycaprolactone composites (SPC) containing 20%, 40%, and 60% silk were prepared by layer-by-layer assembly and hot-pressing technology. Macroscopic morphology was observed and microstructure were observed by scanning electron microscopy, compressive mechanical properties were detected by compression test, surface wettability was detected by surface contact angle test, degradation of materials was observed after soaking in PBS for 180 days, and proliferation of MC3T3-E1 cells was detected by cell counting kit 8 assay. Six Sprague Dawley rats were subcutaneously implanted with polycaprolactone (PCL) and 20%-SPC, respectively. Masson staining was used to analyze the in vivo degradation behavior and vascularization effect within 180 days. Results: The pore defects of the three SPC sections were relatively few. In the range of 20% to 60%, as the silk content increased and the PCL content decreased, the interlayer spacing of silk fabric decreased, and the fibers almost covered the entire cross-section. The compressive modulus and compressive strength of SPC showed an increasing trend, and the compressive modulus of 60%-SPC was slightly lower than that of 40%-SPC. There were significant differences in compressive modulus and compressive strength between the materials ( P<0.05). In vitro simulated fluid degradation experiments showed that the mass loss of the three types of SPC after 180 days of degradation was within 5%, with the highest mass loss observed in 60%-SPC. The differences in mass loss between the materials were significant ( P<0.05). As the silk content increased, the static water contact angle of each material gradually decreased, and all could promote the proliferation of MC3T3-E1 cells. The subcutaneous degradation experiment in rats showed that 20%-SPC began to degrade at 30 days after implantation, and material degradation and vascularization were significant at 180 days, which was in sharp contrast to PCL. Conclusion: SPC has the mechanical and hydrophilic properties that are compatible with bone tissue. It maintains its mechanical strength for a long time in a simulated body fluid environment in vitro, and achieves dynamic synchronization of material degradation, tissue regeneration, and vascularization through the body's immune regulation mechanism in vivo. It is expected to provide a new type of implant material for clinical bone repair.


Assuntos
Teste de Materiais , Poliésteres , Ratos Sprague-Dawley , Seda , Engenharia Tecidual , Poliésteres/química , Animais , Engenharia Tecidual/métodos , Ratos , Seda/química , Camundongos , Substitutos Ósseos/química , Alicerces Teciduais/química , Materiais Biocompatíveis/química , Proliferação de Células , Masculino , Osteoblastos/citologia , Propriedades de Superfície , Osso e Ossos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA