Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Mater Sci Mater Med ; 34(7): 36, 2023 Jul 24.
Article in English | MEDLINE | ID: mdl-37486393

ABSTRACT

The design of hierarchical porous structure in scaffolds is crucial for bone defect regenerative repair. However, bioceramic materials present a challenge in precisely constructing designed micropores owing to the limitation of forming process. To investigate micropore shape influences bone regeneration in bioceramic scaffolds with macropores, hierarchical porous scaffolds with interconnective macropores (~400 µm) and two types of micropores (spherical and fibrous) were prepared using a combination of direct ink writing (DIW) and template sacrifice methods. Compared to the scaffold with spherical micropores, the scaffold with highly interconnected fibrous micropores significantly improved cell adhesion and upregulated osteogenic and angiogenetic-related gene expression in mBMSCs and HUVECs, respectively. Furthermore, in vivo implantation experiments showed that hierarchical scaffolds with fibrous micropores accelerated the bone repair process significantly. This result can be attributed to the high interconnectivity of fibrous micropores, which promotes the transportation of nutrients and waste during bone regeneration. Our work demonstrates that hierarchical porous scaffold design, especially one with a fibrous micropore structure, is a promising strategy for improving the bone regeneration performance of bioceramic scaffolds.


Subject(s)
Bone Regeneration , Tissue Scaffolds , Tissue Scaffolds/chemistry , Calcium Phosphates/chemistry , Osteogenesis , Porosity
2.
Bioact Mater ; 26: 413-424, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36969106

ABSTRACT

Bone bionics and structural engineering have sparked a broad interest in optimizing artificial scaffolds for better bone regeneration. However, the mechanism behind scaffold pore morphology-regulated bone regeneration remains unclear, making the structure design of scaffolds for bone repair challenging. To address this issue, we have carefully assessed diverse cell behaviors of bone mesenchymal stem cells (BMSCs) on the ß-tricalcium phosphate (ß-TCP) scaffolds with three representative pore morphologies (i.e., cross column, diamond, and gyroid pore unit, respectively). Among the scaffolds, BMSCs on the ß-TCP scaffold with diamond pore unit (designated as D-scaffold) demonstrated enhanced cytoskeletal forces, elongated nucleus, faster cell mobility, and better osteogenic differentiation potential (for example, the alkaline phosphatase expression level in D-scaffold were 1.5-2 times higher than other groups). RNA-sequencing analysis and signaling pathway intervention revealed that Ras homolog gene family A (RhoA)/Rho-associated kinase-2 (ROCK2) has in-depth participated in the pore morphology-mediated BMSCs behaviors, indicating an important role of mechanical signaling transduction in scaffold-cell interactions. Finally, femoral condyle defect repair results showed that D-scaffold could effectively promote endogenous bone regeneration, of which the osteogenesis rate was 1.2-1.8 times higher than the other groups. Overall, this work provides insights into pore morphology-mediated bone regeneration mechanisms for developing novel bioadaptive scaffold designs.

3.
J Orthop Translat ; 37: 152-162, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36380884

ABSTRACT

Background: /Objective: Tissue engineering involves scaffolds, cells and growth factors, among which growth factors have limited applications due to potential safety risks and high costs. Therefore, an alternative approach to exogenously induce osteogenesis is desirable. Considering that osteogenesis and angiogenesis are coupled, a system of human umbilical vein endothelial cells (HUVECs) and human bone mesenchymal stem cells (hBMSCs) coculture is more biologically adapted to the microenvironment in vivo and can mediate osteogenesis and angiogenesis via paracrine signalling. Hence, in this study, a HUVECs/hBMSCs coculture system with appropriate cell and medium proportions was established. The substrate for the coculture system was a 3D-printed composite bioceramic scaffold (ß-TCP/CaSiO3) based on a previous study. The aim of this study was to explore the potential of this system for bone tissue engineering. Methods: Bioactive ceramic scaffolds for tissue engineering were fabricated via a 3D Bioplotter™ system. The coculture system for in vitro and in vivo studies consisted of direct contact between HUVECs and hBMSCs cultured on the 3D-printed scaffolds. Results: The proportions of HUVECs/hBMSCs and medium components were determined by cell viability, and the coculture system showed negligible cytotoxicity. CD31 secreted by HUVECs formed strings, and cells tended to aggregate in island chain-like arrays. Real-time cell tracking showed that HUVECs were recruited by hBMSCs, and the integrin expression by HUVECs was upregulated. Ultimately, osteogenic and angiogenic marker gene expression and protein secretion were upregulated. Moreover, the obtained bone tissue engineering scaffolds could induce early osteogenic protein secretion and capillary tube formation in nude rats. Conclusion: These bone tissue engineering scaffolds without exogenous growth factors exhibited the ability to promote osteogenesis/angiogenesis. Translational potential of this article: The fabricated 3D-printed bioactive ceramic scaffolds could provide mechanical, biodegradable and bioadaptive support for personalized bone regeneration. In addition, the bone tissue engineering scaffolds exhibited the ability to promote osteogenesis/angiogenesis without the addition of exogenous growth factors, thus mitigating safety risks. Although application of the HUVECs/hBMSCs coculture system might be a time-consuming process, further development of cord blood storage could be beneficial for multicell coculture.

4.
Bioact Mater ; 12: 120-132, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35087968

ABSTRACT

Calcium phosphate bio-ceramics are osteo-conductive, but it remains a challenge to promote the induction of bone augmentation and capillary formation. The surface micro/nano-topography of materials can be recognized by cells and then the cell fate are mediated. Traditional regulation methods of carving surface structures on bio-ceramics employ mineral reagents and organic additives, which might introduce impurity phases and affect the biological results. In a previous study, a facile and novel method was utilized with ultrapure water as the unique reagent for hydrothermal treatment, and a uniform hydroxyapatite (HAp) surface layer was constructed on composite ceramics (ß-TCP/CaSiO3) in situ. Further combined with 3D printing technology, biomimetic hierarchical structure scaffolds were fabricated with interconnected porous composite ceramic scaffolds as the architecture and micro/nano-rod hybrid HAp as the surface layer. The obtained HAp surface layer favoured cell adhesion, alleviated the cytotoxicity of precursor scaffolds, and upregulated the cellular differentiation of mBMSCs and gene expression of HUVECs in vitro. In vivo studies showed that capillary formation, bone augmentation and new bone matrix formation were upregulated after the HAp surface layer was obtained, and the results confirmed that the fabricated biomimetic hierarchical structure scaffold could be an effective candidate for bone regeneration.

5.
J Mater Chem B ; 9(16): 3573-3583, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33909742

ABSTRACT

The regenerative repair of large bone defects is a major problem in orthopedics and clinical medicine. The key problem is the lack of ability of existing bone graft materials to promote osteogenesis and angiogenesis. Previous studies have shown that the osteogenic or angiogenic abilities of these materials could be significantly improved by adding miRNA or small-molecule drugs to bone graft materials; however, the synergistic effect arising from this combination is not clear. Therefore, we proposed to construct a dual drug delivery system that could simultaneously achieve the co-encapsulation and co-delivery of miRNA and small-molecule drugs to explore the effect of a dual drug delivery system on bone repair. In this study, we constructed dual-sized pore structure calcium-silicon nanospheres (DPNPs) and achieved the co-encapsulation of miR-210, angiogenic gene drugs, and simvastatin (Siv), a small-molecule osteogenic drug, through metal-ion coordination and physical adsorption. In vitro and in vivo osteogenic and angiogenic experiments showed that the dual drug delivery system (Siv/DPNP/miR-210) exhibited better properties than those of the individual unloaded and single drug-loaded systems and could significantly accelerate the process of bone repair, which provides a novel strategy for the regeneration and repair of bone defects.


Subject(s)
Bone Regeneration/drug effects , Drug Delivery Systems , MicroRNAs/metabolism , Simvastatin/pharmacology , Tissue Scaffolds/chemistry , Animals , Calcium/chemistry , Cells, Cultured , Humans , Mice , MicroRNAs/genetics , Nanoparticles/chemistry , Osteogenesis/drug effects , Particle Size , Porosity , Silicon/chemistry , Simvastatin/chemistry , Surface Properties
6.
J Mater Chem B ; 8(23): 5042-5051, 2020 06 21.
Article in English | MEDLINE | ID: mdl-32396152

ABSTRACT

In this study, chitosan-silica hybrids (CSHs) with superior mechanical strength and homogeneous dispersion of nano-sized silica particles were synthesized via a facile sol-gel method aiming for bone regeneration. The effects of varied acidic conditions of sol-gel reaction and inorganic/organic ratios on the performance of the hybrid were investigated. CSHs synthesized under weak acidic conditions (acetic acid, pH 4.0) showed a homogeneous nanostructure and robust strength (maximum compressive strength: 42.6 ± 3.3 MPa and 271 ± 31 MPa in wet and dry forms, respectively). However, those developed under the strong acidic condition (HCl, pH 4.0) and the strong acid condition plus lower pH (HCl, pH 2.8) tended to aggregate and exhibited inferior mechanical properties (compressive strength: 6.3 ± 0.3 MPa in wet form at pH 2.8). Under the latter conditions, the interactions between silica and chitosan were weak. Moreover, the mechanical properties of the CSHs could be tuned in a wide range by conveniently varying the inorganic/organic composition ratio between 50% and 70%. In vitro cytocompatibility study indicated that CSHs were non-cytotoxic. These results suggested that the weak acidic sol-gel process were essential for fabricating chitosan-silica hybrids with high mechanical strength, which had potential to be applied as a bone substitute.


Subject(s)
Bone Regeneration/drug effects , Bone Substitutes/pharmacology , Chitosan/pharmacology , Nanostructures/chemistry , Silicon Dioxide/pharmacology , Tissue Scaffolds/chemistry , Animals , Bone Substitutes/chemistry , Cells, Cultured , Chitosan/chemistry , Mice , Particle Size , Silicon Dioxide/chemistry , Surface Properties
7.
ACS Appl Mater Interfaces ; 10(49): 42146-42154, 2018 Dec 12.
Article in English | MEDLINE | ID: mdl-30507136

ABSTRACT

Titanium (Ti) alloy implants can repair bone defects at load-bearing sites. However, they mechanically mismatch with the natural bone and lack customized adaption with the irregularly major-sized load-bearing bone defects, resulting in the failure of implant fixation. Mineralized collagen (MC), a building block in bone, can induce angiogenesis and osteogenesis, and 3D printing technology can be employed to prepare scaffolds with an overall shape customized to the bone defect. Hence, we induced the formation of MC, made of hydroxyapatite (HAp) nanocrystals and collagen fibers, in 3D-printed porous Ti6Al4V (PT) scaffolds through in situ biomimetic mineralization. The resultant MC/PT scaffolds exhibited a bone-like Young's modulus and were customized to the anatomical contour of actual bone defects of rabbit model. We found that the biocompatibility and osteogenic differentiation are best when the mass ratio between HAp nanocrystals and collagen fibers is 1 in MC. We then implanted the MC/PT scaffolds into the customized radius defect rabbit model and found that the MC/PT scaffolds significantly improved the vascularized bone tissue formation and integration between new bone and the implants. Therefore, a combination of 3D printing and biomimetic mineralization could lead to customized 3D PT scaffolds for enhanced angiogenesis, osteogenesis, and osteointegration. Such scaffolds represent novel patient-specific implants for precisely repairing irregular major-sized load-bearing bone defects.


Subject(s)
Biocompatible Materials , Biomimetic Materials , Calcification, Physiologic/drug effects , Neovascularization, Physiologic/drug effects , Osseointegration/drug effects , Osteogenesis/drug effects , Printing, Three-Dimensional , Radius Fractures , Alloys , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Female , Male , Porosity , Rabbits , Radius Fractures/metabolism , Radius Fractures/pathology , Radius Fractures/therapy , Tissue Scaffolds/chemistry , Titanium/chemistry , Titanium/pharmacology
8.
Carbohydr Polym ; 197: 183-193, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30007604

ABSTRACT

Sol-gel derived organic/inorganic hybrids, in which organic and inorganic components form co-networks at the molecular level, have demonstrated great potential for providing improved mechanical properties and biological functions in tissue engineering applications. Here, a novel bioactive hydroxyapatite-chitosan-silica hybrid (HA-CSH) scaffold was successfully fabricated by combining the sol-gel method and 3D plotting technique. Physiochemical characterization confirmed that chitosan was hybridized homogeneously with the inorganic phase on nanoscale. The obtained scaffolds possessed precisely controllable and interconnected porous structures. The nano-sized HA formed in situ and dispersed uniformly in the hybrid network, which reduced the water absorption and increased the mechanical strength of the hybrid scaffold under humidity condition as compared to chitosan-silica hybrid (CSH) scaffold. Compression tests showed that the 3D plotted hybrid scaffolds under wet conditions had compressive strengths of 10-13 MPa and elastic moduli of 21-27 MPa and thus met the mechanical requirements of human trabecular bone. Studies on the mineralization process under simulated body fluid (SBF) conditions confirmed that the introduction of HA obviously increased the biological activity of hybrid scaffolds. In vitro cell results indicated that the HA-CSH scaffold not only supported adhesion and proliferation of mouse bone mesenchymal stem cells (mBMSCs), but also improved the osteoinductivity. The alkaline phosphatase activity and mineral deposition on the HA-CSH scaffold were higher than those on the CSH scaffold. These results suggested that the 3D plotted HA-CSH scaffold may be a promising bioactive material for bone tissues regeneration.


Subject(s)
Biocompatible Materials/chemical synthesis , Chitosan/chemistry , Durapatite/chemistry , Silicon Dioxide/chemistry , Software , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Bone Regeneration , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chitosan/pharmacology , Durapatite/pharmacology , Gels/chemistry , Humans , Mice , Particle Size , Rheology , Silicon Dioxide/pharmacology , Surface Properties , Tissue Engineering
9.
Adv Healthc Mater ; 7(17): e1800441, 2018 09.
Article in English | MEDLINE | ID: mdl-30044555

ABSTRACT

Due to the difficulty in fabricating bioceramic scaffolds with smaller pore sizes by the current 3D printing technique, the effect of smaller pore sizes (below 400 µm) of 3D printed bioceramic scaffolds on the bone regeneration and biomechanical behavior is never studied. Herein beta-tricalcium phosphate (ß-TCP) scaffolds with interconnected smaller pores of three different sizes (100, 250, and 400 µm) are fabricated by 3D plotting. The resultant scaffolds are then implanted into rat critical-sized calvarial defects without any seeded cells. A custom-designed device is developed to investigate the biomechanical properties of the scaffolds after surgical implantation for 4, 8, and 12 weeks. The scaffolds with the 100 µm pore size are found to present the highest maximum load and stiffness, comparable to those of the autogenous bone, after being implanted for 12 weeks. Micro-computed tomography (micro-CT) and histological analysis further indicate that the scaffolds with the 100 µm pore size achieve the highest percentage of new bone ingrowth, which correlates to their best in vivo biomechanical properties. This study demonstrates that tailoring the pore size of ß-TCP scaffolds to a smaller range by 3D-plotting can be a facile and efficient approach to enhanced bone regeneration and biomechanical behaviors in bone repair.


Subject(s)
Bone Regeneration/physiology , Calcium Phosphates/chemistry , Tissue Scaffolds/chemistry , Animals , Bone Substitutes , Cell Proliferation/physiology , Cells, Cultured , Mice , Osteogenesis/physiology , Porosity , Printing, Three-Dimensional , Rats
10.
J Biomed Mater Res A ; 106(5): 1223-1235, 2018 05.
Article in English | MEDLINE | ID: mdl-29314593

ABSTRACT

Inorganic/organic hybrid silica-chitosan (CS) scaffolds have promising potential for bone defect repair, due to the controllable mechanical properties, degradation behavior, and scaffold morphology. However, the precise in vivo immuno-reactivity of silica-CS hybrids with various compositions is still poorly defined. In this study, we fabricated the three-dimensional (3D) interconnected porous chitosan-silica (CS/SiO2 ) and chitosan-silica-hydroxyapatite (CS/SiO2 /HA) hybrids, through sol-gel process and 3D plotting skill, followed by the naturally or freeze drying separately. Scanning electron microscopy demonstrated the hybrids possessed the uniform geometric structure, while, transmission electron microscopy displayed nanoscale silica, or HA nanoparticles dispersed homogeneously in the CS matrix, or CS/silica hybrids. After intramuscular implantation, CS/SiO2 and CS/SiO2 /HA hybrids triggered a local and limited monocyte/macrophage infiltration and myofiber degeneration. Naturally dried CS/SiO2 hybrid provoked a more severe inflammation than the freeze-dried ones. Dendritic cells were attracted to invade into the implants embedded-muscle, but not be activated to prime the adaptive immunity, because the absence of cytotoxic T cells and B cells in muscle received the implants. Fluorescence-activated cell sorting (FACS) analysis indicated the implanted hybrids were incapable to initiate splenocytes activation. Plasma complement C3 enzyme linked immunosorbent assay (ELISA) assay showed the hybrids induced C3 levels increase in early implanting phase, and the subsequent striking decrease. Thus, the present results suggest that, in vivo, 3D plotted porous CS/SiO2 and CS/SiO2 /HA hybrids are relatively biocompatible in vivo, which initiate a localized inflammatory procedure, instead of a systematic immune response. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1223-1235, 2018.


Subject(s)
Chitosan/immunology , Durapatite/immunology , Silicon Dioxide/immunology , Animals , Biocompatible Materials/chemistry , Complement Activation , Complement C3/metabolism , Compressive Strength , Dystrophin/metabolism , Imaging, Three-Dimensional , Inflammation/pathology , Lymphocytes/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Muscles/metabolism , Muscles/pathology , Porosity , Spleen/pathology
11.
Biomater Sci ; 5(4): 800-807, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28272636

ABSTRACT

Osteopontin (OPN) is a key mediator of cell interactions with biomaterials. However, few studies have been dedicated to studying cell adhesion on OPN-adsorbed substrates with controlled charge and wettability. Here, amino-carboxyl (NH2/COOH) and hydroxyl-methyl (OH/CH3) mixed self-assembled monolayers (SAMs) of varying charges and wettability, respectively, were used as controllable model surfaces to study OPN adsorption and subsequent mesenchymal stem cell (MSC) adhesion. The amount of OPN adsorbed onto the NH2/COOH mixed SAMs appeared to monotonically depend on the surface charge, whereas only a moderately hydrophilic surface was conducive to OPN adsorption on OH/CH3 mixed SAMs. The results correlated well with cell spreading on OPN-coated surfaces in a serum-free medium culture. In addition, the OH/CH3 mixed SAMs with moderate wettability tended to promote ß1, ß3, αv and α5 integrins, indicating that wettability may guide cell adhesion by mediating the integrins signaling pathway. This work will have reference value for designing biologically responsive substrate surfaces.


Subject(s)
Biocompatible Materials/chemistry , Mesenchymal Stem Cells/cytology , Osteopontin/chemistry , Adsorption , Amination , Animals , Cell Adhesion , Cells, Cultured , Methylation , Mice , Surface Properties , Wettability
12.
Colloids Surf B Biointerfaces ; 148: 549-556, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27690244

ABSTRACT

Self-assembled monolayers (SAMs) of alkanethiols on gold are highly controllable model substrates and have been employed to mimic the extracellular matrix for cell-related studies. This study aims to systematically explore how surface chemistry influences the adhesion, morphology, proliferation and osteogenic differentiation of mouse mesenchymal stem cells (mMSCs) using various functional groups (-OEG, -CH3, -PO3H2, -OH, -NH2 and -COOH). Surface analysis demonstrated that these functional groups produced a wide range of wettability and charge: -OEG (hydrophilic and moderate iso-electric point (IEP)), -CH3 (strongly hydrophobic and low IEP), -PO3H2 (moderate wettability and low IEP), -OH (hydrophilic and moderate IEP), -NH2 (moderate wettability and high IEP) and -COOH (hydrophilic and low IEP). In terms of cell responses, the effect of wettability may be more influential than charge for these groups. Moreover, compared to -OEG and -CH3 groups, -PO3H2, -OH, -NH2 and -COOH functionalities tended to promote not only cell adhesion, proliferation and osteogenic differentiation but also the expression of αv and ß1 integrins. This finding indicates that the surface chemistry may guide mMSC activities through αv and ß1 integrin signaling pathways. Model surfaces with controllable chemistry may provide insight into biological responses to substrate surfaces that would be useful for the design of biomaterial surfaces.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Extracellular Matrix/chemistry , Mesenchymal Stem Cells/cytology , Adsorption , Animals , Cell Adhesion/physiology , Cell Differentiation/genetics , Cell Line , Extracellular Matrix/metabolism , Gene Expression , Gold/chemistry , Hydrophobic and Hydrophilic Interactions , Integrins/genetics , Integrins/metabolism , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/metabolism , Mice , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/genetics , Osteogenesis/physiology , Photoelectron Spectroscopy , Reverse Transcriptase Polymerase Chain Reaction , Spectroscopy, Fourier Transform Infrared , Surface Properties , Wettability
13.
Regen Biomater ; 3(5): 269-276, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27699057

ABSTRACT

Yeast cells have controllable biosorption on metallic ions during metabolism. However, few studies were dedicated to using yeast-regulated biomimetic mineralization process to control the strontium-doped positions in calcium phosphate microcapsules. In this study, the yeast cells were allowed to pre-adsorb strontium ions metabolically and then served as sacrificing template for the precipitation and calcination of mineral shell. The pre-adsorption enabled the microorganism to enrich of strontium ions into the inner part of the microcapsules, which ensured a slow-release profile of the trace element from the microcapsule. The co-culture with human marrow stromal cells showed that gene expressions of alkaline phosphatase and Collagen-I were promoted. The promotion of osteogenic differentiation was further confirmed in the 3D culture of cell-material complexes. The strategy using living microorganism as 'smart doping apparatus' to control incorporation of trace element into calcium phosphate paved a pathway to new functional materials for hard tissue regeneration.

14.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 50(1): 38-42, 2015 Jan.
Article in Chinese | MEDLINE | ID: mdl-25779074

ABSTRACT

OBJECTIVE: To investigate the effects of 20% fluor-hydroxyapatite (FHA) on proliferation and osteogenic differentiation of human MG63 osteosarcoma cells. METHODS: FHA was prepared by chemical precipitation method, and its structure and surface features were tested by scanning microscope, X-ray diffraction (XRD) and Fourier transform infrared spectroscopy. MG63 cells were cultured and divided into FHA, hydroxyapatite (HA) and control groups (n = 3). The proliferation of the cells was evaluated using methylthiazol tetrazolium (MTT) assay. ALP activity of the cells was assessed. Osteogenic differentiation was evaluated based on the reverse transcription PCR (RT-PCR) of differentiation-related genes, namely, collagen type I (Col I), alkaline phosphatase (ALP), osteocalcin (OCN) and core binding factor α1 (Cbfα1). The data were analyzed statistically by one-way analysis of variance using SPSS 13.0 software. RESULTS: XRD test showed that the main crystalline phase of FHA was similar to that of HA. Absorptance value of cells exposed to FHA(1.87±0.06) measured by MTT was higher than that of the control(1.25±0.02) on the third day(P < 0.05), and there was no statistically significant difference between the cells exposed to FHA and HA(1.84±0.03) (P > 0.05). ALP activity of the cells exposed to FHA(4.62±0.09)was higher than that of the control (1.92 ± 0.05) (P < 0.05). RT-PCR tests showed that compared with the control, FHA up-regulated the expression of Col I, ALP and OCN mRNA, down-regulated the expression of Cbfα1 mRNA. CONCLUSIONS: FHA enhances the proliferation and osteogenic differentiation-related gene expression, and has good biocompatibility.


Subject(s)
Bone Neoplasms/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Durapatite/pharmacology , Osteosarcoma/pathology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Analysis of Variance , Biocompatible Materials , Bone Neoplasms/metabolism , Cell Differentiation/genetics , Cell Proliferation/physiology , Collagen Type I/genetics , Collagen Type I/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Durapatite/chemistry , Humans , Hydroxyapatites , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis , Osteosarcoma/metabolism , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
15.
J Mater Chem B ; 2(30): 4794-4801, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25328680

ABSTRACT

Self-assembled monolayers (SAMs) of alkanethiols on gold have been employed as model substrates to investigate the effects of surface chemistry on cell behavior. However, few studies were dedicated to the substrates with a controlled wettability in studying stem cell fate. Here, mixed hydroxyl (-OH) and methyl (-CH3) terminated SAMs were prepared to form substrates with varying wettability, which were used to study the effects of wettability on the adhesion, spreading, proliferation and osteogenic differentiation of mesenchymal stem cells (MSCs) from human and mouse origins. The numbers of adhered human fetal MSCs (hMSCs) and mouse bone marrow MSCs (mMSCs) were maximized on -OH/-CH3 mixed SAMs with a water contact angle of 40~70° and 70~90°, respectively. Hydrophilic mixed SAMs with a water contact angle of 20~70° also promoted the spreading of both hMSCs and mMSCs. Both hMSCs and mMSCs proliferation was most favored on hydrophilic SAMs with a water contact angle around 70°. In addition, a moderate hydrophilic surface (with a contact angle of 40~90° for hMSCs and 70° for mMSCs) promoted osteogenic differentiation in the presence of biological stimuli. Hydrophilic mixed SAMs with a moderate wettability tended to promote the expression of αvß1 integrin of MSCs, indicating that the tunable wettability of the mixed SAMs may guide osteogenesis through mediating the αvß1 integrin signaling pathway. Our work can direct the design of biomaterials with controllable wettability to promote the adhesion, proliferation and differentiation of MSCs from different sources.

16.
J Mater Chem B ; 2(29): 4703-4710, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-32262282

ABSTRACT

Understanding the shape effect of hydroxyapatite (HAp) microparticles on cellular behavior is important for enabling new kinds of biological and biomedical applications. However, it is still a challenge to prepare HAp microparticles with different shapes but similar physicochemical properties, and then to investigate their relationships with cellular behavior. Herein, we developed a novel, facile route to regulate the morphology of HAp microparticles, and investigated the interaction between the particles and bone marrow mesenchymal stem cells (BMSCs). Our results revealed that the shape of HAp has a strong influence on cellular behavior, and that the sphere-like particles performed better than the rod-like particles. These findings highlight the importance of the shape characteristics of HAp microparticles, and may provide new insights for the utility of HAp-based materials.

17.
Mater Sci Eng C Mater Biol Appl ; 33(3): 1783-8, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23827636

ABSTRACT

Phosphatidylserine (PS) has been demonstrated to promote bone mineralization. It has also been used in bone repairing biomaterials as a functional molecule. However, the effect of PS on mesenchymal stem cells (MSCs) is not clear. In this study, we determined the effect of PS on the osteogenic differentiation of human MSCs (hMSCs) cultured in growth or osteogenic differentiation medium and the role of the ERK1/2 signaling pathway on PS activity. Cytotoxicity of PS was measured by MTT assay in growth medium for 5 days. Cell osteogenic differentiation was evaluated by alkaline phosphatase (ALP) activity analysis, Alizarin Red S staining and real-time PCR assay. Western blotting and ERK blocking assay were used to examine the role of ERK1/2 signaling pathway on PS activity. The results showed no cytotoxicity for the doses of PS administered. For 21 days, 50-100 µM PS increased ALP expression and mineralization of hMSCs. The expression of the osteogenic gene marker, ALP, osteocalcin (OC), and RUNX2 was enhanced by 50 µM PS treatment at day 14. Phospho-ERK was activated by 50 µM PS at 30 min and 1h in growth medium. In osteogenic medium, 50 µM PS extended phospho-ERK activation by osteogenic induction medium from 30 min to 8 h. U0126, an ERK inhibitor, suppressed the ALP expression induced by PS. Our data indicate that the ERK signal is potentially a mediator in the process of osteogenic differentiation of hMSCs induced by PS. PS, as a functional molecule, has high potential for use in bone repairing biomaterials and bone tissue engineering.


Subject(s)
Cell Differentiation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Osteogenesis/drug effects , Phosphatidylserines/pharmacology , Alkaline Phosphatase/metabolism , Butadienes/pharmacology , Calcification, Physiologic/drug effects , Cell Death/drug effects , Cell Differentiation/genetics , Cells, Cultured , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Mesenchymal Stem Cells/drug effects , Nitriles/pharmacology , Osteogenesis/genetics , Staining and Labeling
18.
Biomed Mater ; 5(5): 054103, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20876955

ABSTRACT

Sol-gel-derived bioactive glasses (BGs) have been developed for bone tissue regeneration. To develop more reliable bone tissue repair systems, it is necessary to control the morphology and surface textures of bioactive glasses. In this study, we prepared bioactive glasses by sol-gel technology using hydrochloride acid, lactic acid, citric acid and acetic acid as hydrolysis catalysts. We studied effects of acids on the morphology and surface textures, apatite-forming bioactivity and cellular response (cellular attachment and proliferation) of BGs. Results showed that the surface morphology, structure, apatite-forming bioactivity and cellular response of BG particles can be controlled by changing acid species. The hydrochloric acid-derived bioactive glass (HBG) and the acetic acid-derived bioactive glass (ABG) present high surface areas and fast apatite-forming rates. Lactic acid- and citric acid-derived bioactive glasses (LBG, CBG) exhibited nanoscale surface morphology, relatively low surface areas and comparable apatite-forming bioactivity. The results of human marrow mesenchymal stem cell (HMSC) culture exhibited that LBG and CBG have an enhanced effect on the cell proliferation, as compared to HBG, ABG and tissue culture plate. This study suggests that sol-gel bioactive glasses with proper surface textures and apatite-forming rate can affect preliminary cellular proliferation.


Subject(s)
Acetic Acid/chemistry , Biocompatible Materials/chemistry , Citric Acid/chemistry , Glass/chemistry , Lactic Acid/chemistry , Bone Marrow Cells/cytology , Bone and Bones/physiology , Catalysis , Cell Adhesion , Cell Proliferation , Cell Survival , Cells, Cultured , Durapatite/chemistry , Gels , Humans , Hydrochloric Acid/chemistry , Hydrolysis , Kinetics , Mesenchymal Stem Cells/physiology , Microscopy, Electron, Scanning , Particle Size , Porosity , Surface Properties
19.
J Biomed Mater Res A ; 94(4): 1091-9, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20694976

ABSTRACT

Bioactive glasses (BGs) have been widely used for bone tissue regeneration as they are able to bond directly with bone. Clinical applications of these materials are likely to be in particulate form. Nanoscale materials can mimic the surface properties of natural tissues, which have exhibited superior cytocompatible property and improved tissue regeneration. The objective of this study is to prepare bioactive glass particles with nanoscale or non-nanoscale surface features and investigate their microstructure, apatite-forming bioactivity and cellular response. The microstructure and micro-nanoscale surface morphology were controlled by adding a hydroxyl-carboxyl acid (citric acid) in the sol-gel process. Results shown that the addition of citric acid induced the formation of nanoscale surface structure and increased the specific surface area, pore volume and pore size of bioactive glass particles. The citric acid with low-concentration-derived sol-gel bioactive glasses (CBGs) resulted in an enhanced apatite-formation ability in simulated body fluids (SBF) compared to normal bioactive glasses. The attachment and proliferation of rat marrow mesenchymal stem cells (RMSCs) on CBGs (low concentration) were higher than those of normal BGs, demonstrating that the CBGs had the excellent cytocompatibility. RMSCs on CBGs (low concentration) expressed the higher alkaline phosphatase activity (ALP) than normal BGs and tissue culture plastic, revealing that CBGs can induced differentiation of RMSCs to the osteogenic lineage. Such improved physical and biological properties of CBGs (low concentration) should be useful in developing new bioactive glass materials for stem cell-based bone regeneration or biomimic tissue engineering scaffolds.


Subject(s)
Cell Differentiation , Glass/chemistry , Mesenchymal Stem Cells/cytology , Nanoparticles/chemistry , Nanotechnology/methods , Adsorption/drug effects , Alkaline Phosphatase/metabolism , Animals , Apatites/pharmacology , Body Fluids , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Citric Acid/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Microscopy, Fluorescence , Nanoparticles/ultrastructure , Nitrogen/metabolism , Particle Size , Porosity/drug effects , Proteins/metabolism , Rats , Surface Properties/drug effects , Temperature , X-Ray Diffraction
20.
Acta Biomater ; 5(5): 1697-707, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19217361

ABSTRACT

Tissue engineering scaffolds with a micro- or nanoporous structure and able to deliver special drugs have already been confirmed to be effective in bone repair. In this paper, we first evaluated the biomineralization properties and drug release properties of a novel mesoporous silica-hydroxyapatite composite material (HMS-HA) which was used as drug vehicle and filler for polymer matrices. Biomineralization can offer a credible prediction of bioactivity for the synthetic bone regeneration materials. We found HMS-HA exhibited good apatite deposition properties after being soaked in simulated body fluid (SBF) for 7 days. Drug delivery from HMS-HA particle was in line with Fick's law, and the release process lasted 12 h after an initial burst release with 60% drug release. A novel tissue engineering scaffold with the function of controlled drug delivery was developed, which was based on HMS-HA particles, poly(lactide-co-glycolide) (PLGA) and microspheres sintering techniques. Mechanical testing on compression, degradation behavior, pH-compensation effect and drug delivery behavior of PLGA/HMS-HA microspheres sintered scaffolds were analyzed. Cell toxicity and cell proliferation on the scaffolds was also evaluated. The results indicated that the PLGA/HMS-HA scaffolds could effectively compensate the increased pH values caused by the acidic degradation product of PLGA. The compressive strength and modulus of PLGA/HMS-HA scaffolds were remarkably high compared to pure PLGA scaffold. Drug delivery testing of the PLGA/HMS-HA scaffolds indicated that PLGA slowed gentamycin sulfate (GS) release from HMS-HA particles, and the release lasted for nearly one month. Adding HMS-HA to PLGA scaffolds improved cytocompatibility. The scaffolds demonstrated low cytotoxicity, and supported mesenchymal stem cells growth more effectively than pure PLGA scaffolds. To summarize, the data supports the development of PLGA/HMS-HA scaffolds as potential degradable and drug delivery materials for bone replacement.


Subject(s)
Bone and Bones/drug effects , Bone and Bones/pathology , Drug Carriers/chemistry , Gentamicins/pharmacology , Silicon Dioxide/chemistry , Tissue Scaffolds , Wound Healing/drug effects , Animals , Body Fluids , Calcification, Physiologic/drug effects , Calcium/chemistry , Cell Death/drug effects , Cell Proliferation/drug effects , Durapatite/chemistry , Hydrogen-Ion Concentration/drug effects , Lactic Acid/chemistry , Mechanical Phenomena/drug effects , Phosphates/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Porosity/drug effects , Solutions , Surface Properties/drug effects , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL
...