Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Bioeng Biotechnol ; 12: 1408702, 2024.
Article in English | MEDLINE | ID: mdl-38978719

ABSTRACT

The incorporation of bioactive ions into biomaterials has gained significant attention as a strategy to enhance bone tissue regeneration on the molecular level. However, little knowledge exists about the effects of the addition of these ions on the immune response and especially on the most important cellular regulators, the macrophages. Thus, this study aimed to investigate the in vitro cytocompatibility and in vivo regulation of bone remodeling and material-related immune responses of a biphasic bone substitute (BBS) coated with metal ions (Sr2+/Mg2+) and PLGA, using the pure BBS as control group. Initially, two cytocompatible modified material variants were identified according to the in vitro results obtained following the DIN EN ISO 10993-5 protocol. The surface structure and ion release of both materials were characterized using SEM-EDX and ICP-OES. The materials were then implanted into Wistar rats for 10, 30, and 90 days using a cranial defect model. Histopathological and histomorphometrical analyses were applied to evaluate material degradation, bone regeneration, osteoconductivity, and immune response. The findings revealed that in all study groups comparable new bone formation were found. However, during the early implantation period, the BBS_Sr2+ group exhibited significantly faster regeneration compared to the other two groups. Additionally, all materials induced comparable tissue and immune responses involving high numbers of both pro-inflammatory macrophages and multinucleated giant cells (MNGCs). In conclusion, this study delved into the repercussions of therapeutic ion doping on bone regeneration patterns and inflammatory responses, offering insights for the advancement of a new generation of biphasic calcium phosphate materials with potential clinical applicability.

2.
Adv Sci (Weinh) ; : e2400242, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874525

ABSTRACT

Maxillofacial bone defects exhibit intricate anatomy and irregular morphology, presenting challenges for effective treatment. This study aimed to address these challenges by developing an injectable bioactive composite microsphere, termed D-P-Ak (polydopamine-PLGA-akermanite), designed to fit within the defect site while minimizing injury. The D-P-Ak microspheres biodegraded gradually, releasing calcium, magnesium, and silicon ions, which, notably, not only directly stimulated the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) but also activated sensory nerve cells to secrete calcitonin gene-related peptide (CGRP), a key factor in bone repair. Moreover, the released CGRP enhanced the osteogenic differentiation of BMSCs through epigenetic methylation modification. Specifically, inhibition of EZH2 and enhancement of KDM6A reduced the trimethylation level of histone 3 at lysine 27 (H3K27), thereby activating the transcription of osteogenic genes such as Runx2 and Osx. The efficacy of the bioactive microspheres in bone repair is validated in a rat mandibular defect model, demonstrating that peripheral nerve response facilitates bone regeneration through epigenetic modification. These findings illuminated a novel strategy for constructing neuroactive osteo-inductive biomaterials with potential for further clinical applications.

3.
Colloids Surf B Biointerfaces ; 236: 113805, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38422666

ABSTRACT

Bone implantation inevitably causes damage to surrounding vasculature, resulting in a hypoxic microenvironment that hinders bone regeneration. Although titanium (Ti)-based devices are widely used as bone implants, their inherent bioinert surface leads to poor osteointegration. Herein, a strontium peroxide (SrO2)-decorated Ti implant, Ti_P@SrO2, was constructed through coating with poly-L-lactic acid (PLLA) to alleviate the hypoxic microenvironment and transform the bioinert surface of the implant into a bioactive surface. PLLA degradation resulted in an acidic microenvironment and the release of SrO2 nanoparticles. The acidic microenvironment then accelerated the decomposition of SrO2, resulting in the release of O2 and Sr ions. O2 released from Ti_P@SrO2 can alleviate the hypoxic microenvironment, thus enhancing cell proliferation in an O2-insufficient microenvironment. Furthermore, under hypoxic and normal microenvironments, Ti_P@SrO2 enhanced alkaline phosphatase activity and bone-related gene expression in C3H10T1/2 cells with the continuous release of Sr ions. Meanwhile, Ti_P@SrO2 suppressed M1 polarization and promoted M2 polarization of bone marrow-derived monocytes under hypoxic and normal conditions. Furthermore, in a rat implantation model, the implant enhanced new bone formation and improved osteointegration after modification with SrO2. In summary, the newly designed O2- and Sr ion-releasing Ti implants are promising for applications in bone defects.


Subject(s)
Prostheses and Implants , Titanium , Animals , Rats , Titanium/pharmacology , Bone Regeneration , Bone and Bones , Ions , Osteogenesis , Surface Properties , Strontium/pharmacology , Osseointegration
4.
Mater Today Bio ; 22: 100749, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37545569

ABSTRACT

Facilitating regeneration of the tendon-to-bone interface can reduce the risk of postoperative retear after rotator cuff repair. Unfortunately, undesirable inflammatory responses following injury, difficulties in fibrocartilage regeneration, and bone loss in the surrounding area are major contributors to suboptimal tendon-bone healing. Thus, the development of biomaterials capable of regulating macrophage polarization to a favorable phenotype and promoting the synchronous regeneration of the tendon-to-bone interface is currently a top priority. Here, strontium-doped mesoporous bioglass nanoparticles (Sr-MBG) were synthesized through a modulated sol-gel method and Bi-lineage Inducible and Immunoregulatory Electrospun Fibers Scaffolds (BIIEFS) containing Sr-MBG were fabricated. The BIIEFS were biocompatible, showed sustained release of multiple types of bioactive ions, enhanced osteogenic and chondrogenic differentiation of mesenchymal stem cells (MSCs), and facilitated macrophage polarization towards the M2 phenotype in vitro. The implantation of BIIEFS at the torn rotator cuff resulted in greater numbers of M2 macrophages and the synchronous regeneration of tendon, fibrocartilage, and bone at the tendon-to-bone interface, leading to a significant improvement in the biomechanical strength of the supraspinatus tendon-humerus complexes. Our research offers a feasible strategy to fabricate immunoregulatory and multi-lineage inducible electrospun fibers scaffolds incorporating bioglass nanoparticles for the regeneration of soft-to-hard tissue interfaces.

5.
Adv Healthc Mater ; 12(25): e2300469, 2023 10.
Article in English | MEDLINE | ID: mdl-37462929

ABSTRACT

The healing of infected bone defects (IBD) is a complex physiological process involving a series of spatially and temporally overlapping events, including pathogen clearance, immunological modulation, vascularization, and osteogenesis. Based on the theory that bone healing is regulated by both biochemical and biophysical signals, in this study, a copper doped bioglass (CuBGs)/methacryloyl-modified gelatin nanoparticle (MA-GNPs)/methacrylated silk fibroin (SilMA) hybrid hydrogel is developed to promote IBD healing. This hybrid hydrogel demonstrates a dual-photocrosslinked interpenetrating network mechanism, wherein the photocrosslinked SilMA as the main network ensures structural integrity, and the photocrosslinked MA-GNPs colloidal network increases strength and dissipates loading forces. In an IBD model, the hydrogel exhibits excellent biophysical characteristics, such as adhesion, adaptation to irregular defect shapes, and in situ physical reinforcement. At the same time, by sequentially releasing bioactive ions such as Cu2+ , Ca2+ , and Si2+ ions from CuBGs on demand, the hydrogel spatiotemporally coordinates antibacterial, immunomodulatory and bone remodeling events, efficiently removing infection and accelerating bone repair without the use of antibiotics or exogenous recombinant proteins. Therefore, the hybrid hydrogel can be used as a simple and effective method for the treatment of IBD.


Subject(s)
Fibroins , Hydrogels , Hydrogels/chemistry , Wound Healing , Osteogenesis , Fibroins/chemistry , Ions/pharmacology
6.
Biomater Adv ; 141: 213120, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36122428

ABSTRACT

Calcium phosphate cement (CPC), a popular injectable bone defect repairing material, has deficiencies in stimulating osteogenesis and angiogenesis. To overcome the weaknesses of CPC, zinc-doped calcium silicate (Zn-CS) which can release bioactive silicon (Si) and zinc (Zn) ions was introduced to CPC. The physicochemical and biological properties of CPC and its composites were evaluated. Firstly, the most effective addition content of calcium silicate (CaSiO3, CS) in promoting the in vitro osteogenesis was first sorted out. On this basis, the most effective Zn doping content in CS for improving osteogenic differentiation of CPC-based composites was screened out. Finally, the immunoregulation of CS/CPC and Zn-CS/CPC in promoting angiogenesis and osteogenesis was studied. The results showed that the most effective incorporation content of CS was 10 wt%. Zn at a doping content of 30 mol% in CS (30Zn-CS) further enhanced the osteogenic capacity of CS/CPC and simultaneously maintained excellent proangiogenic activity. CS/CPC and 30Zn-CS/CPC promoted the recruitment of macrophages and enhanced M2 polarization while inhibiting M1 polarization, which was beneficial to the early vascularization as well as subsequent new bone formation. When implanted into the femoral condylar defects of rabbits, 30Zn-CS/CPC showed high in vivo materials degradation rate, angiogenesis and osteogenesis, due to the synergistic effects of Si and Zn on bio-stimulation and immunoregulation. This study shed light on the synergistic effects of Si and Zn on regulating the angiogenic, osteogenic, and immunoregulatory activity, and 30Zn-CS/CPC is expected to repair the lacunar bone defects effectively.


Subject(s)
Osteogenesis , Zinc , Animals , Bone Cements/pharmacology , Bone Regeneration , Calcium Compounds , Calcium Phosphates/pharmacology , Glass Ionomer Cements/pharmacology , Ions/pharmacology , Rabbits , Silicates , Silicon/pharmacology , Zinc/pharmacology
7.
Small Methods ; 6(8): e2200321, 2022 08.
Article in English | MEDLINE | ID: mdl-35775956

ABSTRACT

Rapid bioactive ion exchange is a form of communication that regulates a wide range of biological processes. Despite advances in super-resolution optical microscopy, visualizing ion exchange remains challenging due to the extremely fast nature of these events. Here, a "converting a dynamic event into a static image construction" (CDtSC) strategy is developed that uses the color transformation of a single dichromatic molecular probe to visualize bioactive ion inter-organelle exchange in live cells. As a proof of concept, a reactive sulfur species (RSS) is analyzed at the mitochondria-lysosome contact sites (MLCs). A non-toxic and sensitive probe based on coumarin-hemicyanine structure is designed that responds to RSS localized in both mitochondria and lysosomes while fluorescing different colors. Using this probe, RSS give-and-take at MLCs is visualized, thus providing the first evidence that RSS is involved in inter-organelle contacts and communication. Taken together, the CDtSC provides a strategy to visualize and analyze rapid inter-organelle ion exchange events in live cells at nanometer resolution.


Subject(s)
Lysosomes , Organelles , Cell Physiological Phenomena , Lysosomes/metabolism , Mitochondria , Mitochondrial Membranes , Organelles/chemistry
8.
Biomaterials ; 274: 120895, 2021 07.
Article in English | MEDLINE | ID: mdl-34020269

ABSTRACT

The development of recombinant protein cross-linked injectable hydrogels with good mechanical strength and effective drug loading capacity for bone regeneration is extremely attractive and rarely reported. Here, we report the fabrication of a smart hydrogel delivery system by incorporating a rationally designed T4 lysozyme mutant (T4M) to mediate the localized delivery and synergistic release of Mg2+ and Zn2+ for bone repair. Apart from its intrinsic antibacterial properties, T4M bears abundant free amine groups on its surface to function as effective covalent crosslinkers to strengthen the hydrogel network as well as exhibits specific binding affinity to multivalent cations such as Zn2+. Moreover, the integrin receptor-binding Arg-Gly-Asp (RGD) sequence was introduced onto the C-terminus of T4 lysozyme to improve its cellular affinity and further facilitate rapid tissue regeneration. The final composite hydrogel displays excellent injectability, improved mechanical properties, antibacterial activity, and unique bioactivities. The effective loading of Mg2+/Zn2+ in the hydrogels could mediate the sequential and sustained release of Mg2+ and Zn2+, thereby resulting in synergistic enhancement on bone regeneration through modulation of the MAPK signaling pathway. We believe that the strategy proposed in this paper opens up a new route for developing protein cross-linked smart delivery systems for tissue regeneration.


Subject(s)
Hydrogels , Magnesium , Bone Regeneration , Ions , Zinc
9.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 39(2): 123-128, 2021 Apr 01.
Article in Chinese | MEDLINE | ID: mdl-33834665

ABSTRACT

Oromaxillofacial hard tissue defects is still a difficult problem in clinical treatment. Regeneration of oromaxillofacial hard tissue based on tissue engineering technology has a good clinical application prospect. The functional modification of scaffolds is one of key factors that influence the outcome of tissue regeneration. The biomimetic design of biomaterials through simulating the natural structure and composition of oromaxillofacial hard tissue has gradually become a research hotspot due to its advantages of simplicity and efficiency. In this article, the biomimetic modification of biomaterials for oromaxillofacial hard tissue regeneration is reviewed, expecting to provide a new idea for the treatment of oromaxillofacial hard tissue defect.


Subject(s)
Dental Implants , Tissue Scaffolds , Biocompatible Materials , Biomimetics , Bone Regeneration , Tissue Engineering
10.
Mater Sci Eng C Mater Biol Appl ; 118: 111393, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33254998

ABSTRACT

OssiMend® Bioactive (Collagen Matrix Inc., NJ) is a three-component porous composite bone graft device of 45S5 Bioglass/carbonate apatite/collagen. Our in vitro studies showed that conditioned media of the dissolution products of OssiMend Bioactive stimulated primary human osteoblasts to form mineralized bone-like nodules in vitro in one week, in basal culture media (no osteogenic supplements). Osteoblast differentiation was followed by gene expression analysis and a mineralization assay. In contrast, the dissolution products from commercial OssiMend (Bioglass-free carbonate apatite/collagen scaffolds), or from 45S5 Bioglass particulate alone, did not induce the mineralization of the extracellular matrix, but did induce osteoblast differentiation to mature osteoblasts, evidenced by the strong upregulation of BGLAP and IBSP mRNA levels. The calcium ions and soluble silicon species released from 45S5 Bioglass particles and additional phosphorus release from OssiMend mediated the osteostimulatory effects. Medium conditioned with OssiMend Bioactive dissolution had a much higher concentration of phosphorus and silicon than media conditioned with OssiMend and 45S5 Bioglass alone. While OssiMend and OssiMend Bioactive led to calcium precipitation in cell culture media, OssiMend Bioactive produced a higher concentration of soluble silicon than 45S5 Bioglass and higher dissolution of phosphorus than OssiMend. These in vitro results suggest that adding 45S5 Bioglass to OssiMend produces a synergistic osteostimulation effect on primary human osteoblasts. In summary, dissolution products of a Bioglass/carbonate apatite/collagen composite scaffold (OssiMend® Bioactive) stimulate human osteoblast differentiation and mineralization of extracellular matrix in vitro without any osteogenic supplements. The mineralization was faster than for dissolution products of ordinary Bioglass.


Subject(s)
Biocompatible Materials , Ceramics , Apatites , Cell Differentiation , Ceramics/pharmacology , Collagen , Glass , Humans , Osteoblasts , Solubility
11.
Biomaterials ; 264: 120414, 2021 01.
Article in English | MEDLINE | ID: mdl-32980635

ABSTRACT

The treatment of chronic wounds is a major challenge in regenerative medicine, and angiogenesis is known to be critical for chronic wound healing. Hot springs with temperature in the range of 30-45 °C can promote blood circulation, and some hot spring elements including iron and silicon are also known to be active in promoting angiogenesis. Inspired by the hot spring function, we designed a novel bioactive photothermal hydrogel with "hot spring effect" based on fayalite (FA) and N, O-carboxymethyl chitosan (NOCS), which releases bioactive ions and has heating function to create hot ion environment in wound area. The hot spring-mimetic hydrogel showed significant enhancement of angiogenesis and chronic wound healing in vivo due to the in situ heating through photothermal effect combined with the bioactive ions (Fe2+ and SiO44-) released from the hydrogel. It is further confirmed that the synergetic effect of the mild heating and bioactive ions on angiogenesis was mainly because of the activation of different angiogenic factors and signaling pathways. Our study suggests that the hot spring-mimetic approach may be an effective strategy to design bioactive materials for promoting angiogenesis and tissue regeneration.


Subject(s)
Hot Springs , Hydrogels , Regenerative Medicine , Silicon , Wound Healing
12.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-878420

ABSTRACT

Oromaxillofacial hard tissue defects is still a difficult problem in clinical treatment. Regeneration of oromaxillofacial hard tissue based on tissue engineering technology has a good clinical application prospect. The functional modification of scaffolds is one of key factors that influence the outcome of tissue regeneration. The biomimetic design of biomaterials through simulating the natural structure and composition of oromaxillofacial hard tissue has gradually become a research hotspot due to its advantages of simplicity and efficiency. In this article, the biomimetic modification of biomaterials for oromaxillofacial hard tissue regeneration is reviewed, expecting to provide a new idea for the treatment of oromaxillofacial hard tissue defect.


Subject(s)
Biocompatible Materials , Biomimetics , Bone Regeneration , Dental Implants , Tissue Engineering , Tissue Scaffolds
13.
Chin J Dent Res ; 22(2): 93-104, 2019.
Article in English | MEDLINE | ID: mdl-31172137

ABSTRACT

The repair of large bone defects remains a huge challenge for bone regenerative medicine. To meet this challenge, a number of bone substitutes have been developed over recent years to overcome the drawbacks of traditional autograft and allograft therapies. Thus, the improvement of the osteoinductive ability of these substitutes has become a major focus for research in the field of bone tissue engineering. It has been reported that some metallic ions play an important role in bone metabolism in the human body, and that bone repair could be enhanced by incorporating these ions into bone substitutes. Moreover, it is well documented that ions released from these substitutes such as magnesium, zinc, and strontium can increase the osteogenic and angiogenic properties of bone repair scaffolds. However, the mechanisms of action of these ions on cellular bioactivity are currently unclear. Therefore, in the present article, we highlight the recent use of bioactive ions in bone tissue engineering and discuss the effects of these ions on osteogenesis and neovascularisation.


Subject(s)
Bone Regeneration , Bone Substitutes , Humans , Ions , Osteogenesis , Tissue Engineering , Tissue Scaffolds
14.
Adv Sci (Weinh) ; 6(1): 1801260, 2019 Jan 09.
Article in English | MEDLINE | ID: mdl-30643722

ABSTRACT

Although numerous therapies are widely applied clinically and stem cells and/or biomaterial based in situ implantations have achieved some effects, few of these have observed robust myocardial regeneration. The beneficial effects on cardiac function and structure are largely acting through paracrine signaling, which preserve the border-zone around the infarction, reduce apoptosis, blunt adverse remodeling, and promote angiogenesis. Ionic extracts from biomaterials have been proven to stimulate paracrine effects and promote cell-cell communications. Here, the paracrine stimulatory function of bioactive ions derived from biomaterials is integrated into the clinical concept of administration and proposed "ion therapy" as a novel strategy for myocardial infarction. In vitro, silicon- enriched ion extracts significantly increase cardiomyocyte viability and promote cell-cell communications, thus stimulating vascular formation via a paracrine effect under glucose/oxygen deprived conditions. In vivo, by intravenous injection, the bioactive silicon ions act as "diplomats" and promote crosstalk in myocardial cells, stimulate angiogenesis, and improve cardiac function post-myocardial infarction.

15.
ACS Appl Mater Interfaces ; 11(1): 1449-1468, 2019 Jan 09.
Article in English | MEDLINE | ID: mdl-30543278

ABSTRACT

Cardiac tissue engineering (CTE) remains a great challenge to construct a cell-inductive scaffold that has positive effects on cardiac cell behaviors and cardiac tissue repair. In this study, we for the first time demonstrated that Si ions evidently stimulated the expression of cardiac-specific genes and proliferation of neonatal rat cardiomyocytes (NRCMs) at concentration ranges of 0.13-10.78 ppm. Accordingly, the optimized concentrations of calcium silicate (CS) were incorporated into the controllable aligned chitosan electrospun nanofibers, constructing the composite cardiac patch scaffolds. These scaffolds showed synergistic effect of bioactive chemical and structural signals on both cardiomyocytes and endothelial cells with aligned cell morphology and enhanced viability and function characterized by upregulated expressions of cardiac and angiogenic specific markers, improved myofilament structure, and better Ca2+ transients of NRCMs as compared to the scaffolds free of CS component or with disordered structures. The in vivo studies further demonstrated that the NRCM-seeded aligned CS/chitosan cardiac patch evidently improved cardiac function via limiting the scar area and promoting angiogenesis in postmyocardial infarction rats. Conclusively, our study highlights the potential application of bioactive ions and nanostructured biomaterials in CTE, and the CS/chitosan composite cardiac patch may be a promising scaffold for repair of infarcted myocardium.


Subject(s)
Calcium Compounds , Chitosan , Myocardial Infarction , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Nanofibers/chemistry , Silicates , Animals , Calcium Compounds/chemistry , Calcium Compounds/pharmacology , Cells, Cultured , Chitosan/chemistry , Chitosan/pharmacology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardium/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Silicates/chemistry , Silicates/pharmacology
16.
Acta Biomater ; 66: 81-92, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28864248

ABSTRACT

Silicate bioceramics have been considered to possess a wide prospect of clinical application for orthopedic tissue regeneration due to their excellent osteogenesis and angiogenesis. However, the mechanism for silicate bioceramics stimulating bone formation is not fully understood. The host immune defense to implants is proved to greatly influence the osteogenesis and new bone formation, but up to now, few studies are focused on the silicate bioceramics modulated host immune responses. In our present study, two representative silicate bioceramics, akermanite (AKT) and nagelschmidtite (NAGEL) were used as model materials to investigate the inflammation responses in vitro and in vivo, and ß-tricalcium phosphate (ß-TCP) bioceramics were used as a control. It was found that the mouse macrophage cell RAW264.7 that cultured on AKT and NAGEL bioceramics displayed not only less viability and proliferation, but also a significant less inflammatory cytokine secretion than those on ß-TCP in vitro. The formation of foreign body giant cells and fibrous capsules, the invasion of macrophages, as well as the detected inflammatory cytokines around the implanted materials were much lower in both AKT and NAGEL bioceramic groups as compared with those in the ß-TCP controls in vivo. Furthermore, it was found that not just the certain concentration of extracellular Si-containing ionic products released from the silicate bioceramics, but also the separate Si, Mg and Ca ions revealed the activity to inhibit the macrophage inflammatory responses by the way of suppressing the activated inflammatory MAPK and NF-κB signaling pathway and promoting the caspase-dependent apoptosis of macrophages. In general, our study suggests that the silicate bioceramics could regulate immune responses by altering the ionic microenvironment between the implants and hosts, which may offer new insight about the mechanism of the bioactivity of silicate bioceramics in bone regeneration and provide profitable guidance for designing new biomaterials for bone tissue engineering. STATEMENT OF SIGNIFICANCE: Silicate bioceramics have been widely used for orthopedic tissue regeneration because of their excellent characteristics in bone formation. However, there are few studies concerning their interrelationships with the host immune defense that has been proved to greatly influence osteogenesis. In our present study, the akermanite and nagelschmidtite were used as two representative silicate bioceramics to investigate the inflammation responses in vitro and in vivo; and for the first time, the bioactive ions released from the silicate bioceramics were discovered to regulate the macrophage immune responses through both inhibiting the inflammatory signaling and activating apoptosis of macrophages. Our findings in this study may not only increase the understanding in osteogenic activity of silicate bioceramics, but also provide profitable guidance for designing and manufacturing new biomaterials for bone tissue engineering.


Subject(s)
Biocompatible Materials/pharmacology , Bone Regeneration/drug effects , Ceramics/pharmacology , Immunity/drug effects , Silicates/pharmacology , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/metabolism , Foreign Bodies/immunology , Foreign Bodies/pathology , Implants, Experimental , Ions , Lipopolysaccharides , MAP Kinase Signaling System/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , RAW 264.7 Cells
17.
Acta Biomater ; 61: 217-232, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28807800

ABSTRACT

Bioactive ions released from bioceramics play important roles in bone regeneration; however, it is unclear how each ionic composition in complex bioceramics exerts its specific effect on bone regeneration. The aim of this study is to elucidate the functional effects of Sr and Si ions in bioceramics on the regeneration of osteoporotic bone. A model bioceramic with Sr- and Si-containing components (SMS) was successfully fabricated and the effects of ionic products from SMS bioceramics on the osteogenic, osteoclastic and angiogenic differentiation of rBMSCs-OVX and RANKL-induced osteoclasts were investigated. The results showed that SMS bioceramics could enhance ALP activity and expression of Col 1, OCN, Runx2, and angiogenic factors including VEGF and Ang-1. SMS bioceramics not only rebalanced the OPG/RANKL ratio of rBMSCs-OVX at early stage, but also repressed RANKL-induced osteoclast formation and expression of TRAP, DC-STAMP, V-ATPase a3, and NFATc1. The synergistic effects of Sr and Si ions were further investigated as compared with those of similar concentrations of Sr and Si ions alone. Sr and Si ions possessed synergistic effects on osteogenesis, osteoclastogenesis, and angiogenesis, attributed to the dominant effects of Sr ions on enhancing angiogenesis and repressing osteoclastogenesis, and the dominant effects of Si ions on stimulating osteogenesis. The in vivo study using critical-size mandibular defects of OVX rat models showed that SMS bioceramics could significantly enhance bone formation and mineralization compared with ß-TCP bioceramics. Our results are the first to elucidate the specific effect of each ion from bioceramics on osteogenesis, osteoclastogenesis and angiogenesis for osteoporotic bone regeneration, paving the way for the design of functional biomaterials with complex compositions for tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE: Bioactive ions released from bioceramics play important roles for bone regeneration; however, it is unclear how each of ionic compositions in complex bioceramics exerts its specific effect on bone regeneration. The aim of present study is to elucidate the functional effects of Sr and Si ions in complex bioceramics on the regeneration of osteoporotic bone. A model bioceramic with Sr and Si-containing components (SMS) was successfully fabricated and the effects of ionic products from SMS bioceramics on the osteogenic, osteoclastic and angiogenic differentiation of rBMSCs-OVX and RANKL-induced osteoclasts were investigated. The results showed that SMS bioceramics could enhance ALP activity and expression of Col 1, OCN, Runx2 and angiogenic factors including VEGF and Ang-1. SMS bioceramics not only rebalanced the ratio of OPG/RANKL of OVX-BMSCs at early stage, but also repressed RANKL-induced osteoclast formation and expression of TRAP, DC-STAMP, V-ATPase a3, and NFATc1. The synergistic effects of Sr and Si ions were further investigated as compared with the similar concentration of Sr and Si ions alone. It was found that Sr and Si ions possessed synergistic effects on osteogenesis, osteoclastogenesis and angiogenesis, attributed to the dominant effects of Sr ions on enhancing angiogenesis and repressing osteoclastogenesis, and the dominant effects of Si ions on stimulating osteogenesis. The in vivo study using critical-size mandibular defects of OVX rat models showed that SMS bioceramics could significantly enhance bone formation and mineralization as compared with ß-TCP bioceramics. It is suggested that SMS bioceramics may be a promising biomaterial for osteoporotic bone regeneration. To our knowledge, this is the first time to elucidate the specific effect of each ion from bioceramics on osteogenesis, osteoclastogenesis and angiogenesis for osteoporotic bone regeneration, paving the way to design functional biomaterials with complex compositions for tissue engineering and regenerative medicine.


Subject(s)
Biocompatible Materials/pharmacology , Bone Regeneration/drug effects , Neovascularization, Physiologic/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Osteoporosis/physiopathology , Silicon/pharmacology , Strontium/pharmacology , Alkaline Phosphatase/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Fluorescence , Ions , Mandible/diagnostic imaging , Mandible/pathology , Mice , Osteoclasts/drug effects , Osteoporosis/pathology , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Tartrate-Resistant Acid Phosphatase/metabolism , X-Ray Microtomography
18.
J Biomed Mater Res A ; 102(7): 2096-104, 2014 Jul.
Article in English | MEDLINE | ID: mdl-23894078

ABSTRACT

Porous ß-CaSiO3/ß-Ca3(PO4)2 (ß-CS/ß-TCP) composite scaffolds have been previously shown to promote bone formation in vivo. However, the mechanisms underlying such beneficial effects remain unclear. In this study, we recreated an extracellular environment using the extracts of ß-CS/ß-TCP composites developed in our previous in vivo study, and investigated the effects of the extracts on osteogenic differentiation of rat bone marrow-derived mesenchymal stem cells (rBMSCs) and its related mechanisms. The angiogenic potential of the extracts was also evaluated using human umbilical vein endothelial cells (HUVECs). In the absence of osteogenic supplements, the osteogenic differentiation of rBMSCs was detected by alkaline phosphatase (ALP) activity assay and the messenger RNA expression of a panel of osteoblast markers. The results showed that the soluble ions of porous ß-CS/ß-TCP composites were capable of promoting cell viability, directly inducing cell differentiation. The increase in phosphorylation of AMP-activated protein kinase (AMPK) and ERK1/2 were observed in rBMSCs cultured in ß-CS/ß-TCP composite extracts. The ALP expression, calcium deposition, and ERK1/2 phosphorylation of rBMSCs, which was promoted by ions released from ß-CS/ß-TCP composites, were blocked by an AMPK inhibitor, Compound C. These results indicate that bioactive ions extracted from ß-CS/ß-TCP composites could stimulate the osteogenic differentiation of rBMSCs via the AMPK-Erk1/2 pathway. Interestingly, the secretion of vascular endothelial growth factor and the viability of HUVECs were shown to be enhanced in the presence of extracts from the ß-CS/ß-TCP composite scaffolds. Our findings suggest that 50 or 80% wt. CS could promote bone regeneration by stimulating osteogenesis and angiogenesis.


Subject(s)
Bone and Bones/cytology , Calcium Compounds , Calcium Phosphates , Cell Differentiation , Endothelium, Vascular/metabolism , Silicates , Tissue Scaffolds , Vascular Endothelial Growth Factor A/metabolism , Adenylate Kinase/metabolism , Animals , Base Sequence , DNA Primers , Endothelium, Vascular/cytology , Human Umbilical Vein Endothelial Cells , Humans , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...