Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 602
Filtrar
1.
J Mater Sci Mater Med ; 35(1): 40, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39073605

RESUMO

Bone infections are still a major problem in surgery. To avoid severe side effects of systemically administered antibiotics, local antibiotic therapy is increasingly being considered. Using a pressure-based method developed in our group, microporous ß-TCP ceramics, which had previously been characterized, were loaded with 2% w/v alginate containing 50 mg/mL clindamycin and 10 µg/mL rhBMP-2. Release experiments were then carried out over 28 days with changes of liquid at defined times (1, 2, 3, 6, 9, 14, 21 and 28d). The released concentrations of clindamycin were determined by HPLC and those of rhBMP-2 by ELISA. Continuous release (anomalous transport) of clindamycin and uniform release (Fick's diffusion) of BMP-2 were determined. The composites were biocompatible (live/dead, WST-I and LDH) and the released concentrations were all antimicrobially active against Staph. aureus. The results were very promising and clindamycin was detected in concentrations above the MIC as well as a constant rhBMP-2 release over the entire study period. Biocompatibility was also not impaired by either the antibiotic or the BMP-2. This promising approach can therefore be seen as an alternative to the common treatment with PMMA chains containing gentamycin, as the new composite is completely biodegradable and no second operation is necessary for removal or replacement.


Assuntos
Antibacterianos , Materiais Biocompatíveis , Proteína Morfogenética Óssea 2 , Clindamicina , Staphylococcus aureus , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/administração & dosagem , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/farmacocinética , Clindamicina/administração & dosagem , Clindamicina/química , Clindamicina/farmacocinética , Humanos , Materiais Biocompatíveis/química , Staphylococcus aureus/efeitos dos fármacos , Cinética , Fosfatos de Cálcio/química , Animais , Teste de Materiais , Proteínas Recombinantes/química , Cerâmica/química , Fator de Crescimento Transformador beta , Alginatos/química , Implantes Absorvíveis , Testes de Sensibilidade Microbiana
2.
ACS Appl Mater Interfaces ; 16(30): 39165-39180, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39041490

RESUMO

Methods for promoting and controlling the differentiation of human mesenchymal stem cells (hMSCs) in vitro before in vivo transplantation are crucial for the advancement of tissue engineering and regenerative medicine. In this study, we developed poly(ethylene glycol) diacrylate (PEGDA) hydrogels with tunable mechanical properties, including elasticity and viscoelasticity, coupled with bioactivity achieved through the immobilization of a mixture of RGD and a mimetic peptide of the BMP-2 protein. Despite the key relevance of hydrogel mechanical properties for cell culture, a standard for its characterization has not been proposed, and comparisons between studies are challenging due to the different techniques employed. Here, a comprehensive approach was employed to characterize the elasticity and viscoelasticity of these hydrogels, integrating compression testing, rheology, and atomic force microscopy (AFM) microindentation. Distinct mechanical behaviors were observed across different PEGDA compositions, and some consistent trends across multiple techniques were identified. Using a photoactivated cross-linker, we controlled the functionalization density independently of the mechanical properties. X-ray photoelectrin spectroscopy and fluorescence microscopy were employed to evaluate the functionalization density of the materials before the culturing of hMSCs on them. The cells cultured on all functionalized hydrogels expressed an early osteoblast marker (Runx2) after 2 weeks, even in the absence of a differentiation-inducing medium compared to our controls. Additionally, after only 1 week of culture with osteogenic differentiation medium, cells showed accelerated differentiation, with clear morphological differences observed among cells in the different conditions. Notably, cells on stiff but stress-relaxing hydrogels exhibited an overexpression of the osteocyte marker E11. This suggests that the combination of the functionalization procedure with the mechanical properties of the hydrogel provides a potent approach to promoting the osteogenic differentiation of hMSCs.


Assuntos
Regeneração Óssea , Elasticidade , Hidrogéis , Células-Tronco Mesenquimais , Polietilenoglicóis , Humanos , Polietilenoglicóis/química , Hidrogéis/química , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Regeneração Óssea/efeitos dos fármacos , Viscosidade , Diferenciação Celular/efeitos dos fármacos , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/farmacologia , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Engenharia Tecidual/métodos
3.
BMC Biotechnol ; 24(1): 38, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38831403

RESUMO

BACKGROUND: Antibiotic-containing carrier systems are one option that offers the advantage of releasing active ingredients over a longer period of time. In vitro sustained drug release from a carrier system consisting of microporous ß-TCP ceramic and alginate has been reported in previous works. Alginate dialdehyde (ADA) gelatin gel showed both better mechanical properties when loaded into a ß-TCP ceramic and higher biodegradability than pure alginate. METHODS: Dual release of daptomycin and BMP-2 was measured on days 1, 2, 3, 6, 9, 14, 21, and 28 by HPLC and ELISA. After release, the microbial efficacy of the daptomycin was verified and the biocompatibility of the composite was tested in cell culture. RESULTS: Daptomycin and the model compound FITC protein A (n = 30) were released from the composite over 28 days. A Daptomycin release above the minimum inhibitory concentration (MIC) by day 9 and a burst release of 71.7 ± 5.9% were observed in the loaded ceramics. Low concentrations of BMP-2 were released from the loaded ceramics over 28 days.


Assuntos
Antibacterianos , Proteína Morfogenética Óssea 2 , Fosfatos de Cálcio , Cerâmica , Daptomicina , Gelatina , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/metabolismo , Daptomicina/química , Daptomicina/farmacologia , Gelatina/química , Cerâmica/química , Antibacterianos/química , Antibacterianos/farmacologia , Fosfatos de Cálcio/química , Animais , Testes de Sensibilidade Microbiana , Camundongos , Portadores de Fármacos/química , Liberação Controlada de Fármacos
4.
ACS Appl Bio Mater ; 7(7): 4293-4306, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38917363

RESUMO

The present work aims to develop optimized scaffolds for bone repair by incorporating mesoporous nanoparticles into them, thereby combining bioactive factors for cell growth and preventing rapid release or loss of effectiveness. We synthesized biocompatible and biodegradable scaffolds designed for the controlled codelivery of curcumin (CUR) and recombinant human bone morphogenic protein-2 (rhBMP-2). Active agents in dendritic silica/titania mesoporous nanoparticles (DSTNs) were incorporated at different weight percentages (0, 2, 5, 7, 9, and 10 wt %) into a matrix of polycaprolactone (PCL) and polyethylene glycol (PEG) nanofibers, forming the CUR-BMP-2@DSTNs/PCL-PEG delivery system (S0, S2, S5, S7, S9, and S10, respectively, with the number showing the weight percentage). To enhance the formation process, the system was treated using low-intensity pulsed ultrasound (LIPUS). Different advanced methods were employed to assess the physical, chemical, and mechanical characteristics of the fabricated scaffolds, all confirming that incorporating the nanoparticles improves their mechanical and structural properties. Their hydrophilicity increased by approximately 25%, leading to ca. 53% enhancement in their water absorption capacity. Furthermore, we observed a sustained release of approximately 97% for CUR and 70% for BMP-2 for the S7 (scaffold with 7 wt % DSTNs) over 28 days, which was further enhanced using ultrasound. In vitro studies demonstrated accelerated scaffold biodegradation, with the highest level observed in S7 scaffolds, approximately three times higher than the control group. Moreover, the cell viability and proliferation on DSTNs-containing scaffolds increased when compared to the control group. Overall, our study presents a promising nanocomposite scaffold design with notable improvements in structural, mechanical, and biological properties compared to the control group, along with controlled and sustained drug release capabilities. This makes the scaffold a compelling candidate for advanced bone tissue engineering and regenerative therapies.


Assuntos
Materiais Biocompatíveis , Proteína Morfogenética Óssea 2 , Teste de Materiais , Nanofibras , Osteogênese , Tamanho da Partícula , Osteogênese/efeitos dos fármacos , Nanofibras/química , Proteína Morfogenética Óssea 2/química , Humanos , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Ondas Ultrassônicas , Curcumina/química , Curcumina/farmacologia , Proliferação de Células/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Poliésteres/química , Polímeros/química , Alicerces Teciduais/química , Polietilenoglicóis/química , Fator de Crescimento Transformador beta
5.
J Biomater Appl ; 39(3): 195-206, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38877801

RESUMO

To reduce the risk of nonunion after spinal fusion surgery, the in situ transplantation of bone marrow mesenchymal stem cells (BMSCs) induced toward osteogenic differentiation by bone morphogenetic protein-2 (BMP2) has been proven effective. However, the current biological agents used for transplantation have limitations, such as a short half-life and low bioavailability. To address this, our study utilized a safe and effective gelatin-methacryloyl (GelMA) as a carrier for BMP2. In vitro, experiments were conducted to observe the ability of this composite vehicle to induce osteogenic differentiation of BMSCs. The results showed that the GelMA hydrogel, with its critical properties and controlled release performance of BMP2, exhibited a slow release of BMP2 over 30 days. Moreover, the GelMA hydrogel not only enhanced the proliferation activity of BMSCs but also significantly promoted their osteogenic differentiation ability, surpassing the BMP2 effects. To investigate the potential of the GelMA-BMP2 composite vehicle, a rabbit model was employed to explore its ability to induce in situ intervertebral fusion by BMSCs. Transplantation experiments in rabbits demonstrated the effective induction of intervertebral bone fusion by the GelMA-BMP2-BMSC composite vehicle. In conclusion, the GelMA-BMP2-BMSC composite vehicle shows promising prospects in preclinical translational therapy for spinal intervertebral fusion. It addresses the limitations of current biological agents and offers a controlled release of BMP2, enhancing the proliferation and osteogenic differentiation of BMSCs.


Assuntos
Proteína Morfogenética Óssea 2 , Diferenciação Celular , Gelatina , Células-Tronco Mesenquimais , Metacrilatos , Osteogênese , Fusão Vertebral , Animais , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/administração & dosagem , Proteína Morfogenética Óssea 2/farmacologia , Gelatina/química , Coelhos , Osteogênese/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Metacrilatos/química , Diferenciação Celular/efeitos dos fármacos , Portadores de Fármacos/química , Hidrogéis/química , Transplante de Células-Tronco Mesenquimais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Masculino , Humanos
6.
BMC Biotechnol ; 24(1): 34, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38783306

RESUMO

BACKGROUND: Signal peptide (SP) engineering has proven able to improve production of many proteins yet is a laborious process that still relies on trial and error. mRNA structure around the translational start site is important in translation initiation and has rarely been considered in this context, with recent improvements in in silico mRNA structure potentially rendering it a useful predictive tool for SP selection. Here we attempt to create a method to systematically screen candidate signal peptide sequences in silico based on both their nucleotide and amino acid sequences. Several recently released computational tools were used to predict signal peptide activity (SignalP), localization target (DeepLoc) and predicted mRNA structure (MXFold2). The method was tested with Bone Morphogenetic Protein 2 (BMP2), an osteogenic growth factor used clinically for bone regeneration. It was hoped more effective BMP2 SPs could improve BMP2-based gene therapies and reduce the cost of recombinant BMP2 production. RESULTS: Amino acid sequence analysis indicated 2,611 SPs from the TGF-ß superfamily were predicted to function when attached to BMP2. mRNA structure prediction indicated structures at the translational start site were likely highly variable. The five sequences with the most accessible translational start sites, a codon optimized BMP2 SP variant and the well-established hIL2 SP sequence were taken forward to in vitro testing. The top five candidates showed non-significant improvements in BMP2 secretion in HEK293T cells. All showed reductions in secretion versus the native sequence in C2C12 cells, with several showing large and significant decreases. None of the tested sequences were able to increase alkaline phosphatase activity above background in C2C12s. The codon optimized control sequence and hIL2 SP showed reasonable activity in HEK293T but very poor activity in C2C12. CONCLUSIONS: These results support the use of peptide sequence based in silico tools for basic predictions around signal peptide activity in a synthetic biology context. However, mRNA structure prediction requires improvement before it can produce reliable predictions for this application. The poor activity of the codon optimized BMP2 SP variant in C2C12 emphasizes the importance of codon choice, mRNA structure, and cellular context for SP activity.


Assuntos
Proteína Morfogenética Óssea 2 , Sinais Direcionadores de Proteínas , RNA Mensageiro , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/química , Sinais Direcionadores de Proteínas/genética , Humanos , RNA Mensageiro/genética , RNA Mensageiro/química , Sequência de Aminoácidos , Conformação de Ácido Nucleico , Biologia Computacional/métodos , Engenharia de Proteínas/métodos , Células HEK293
7.
ACS Biomater Sci Eng ; 10(4): 2414-2425, 2024 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-38446137

RESUMO

Bone defects are a common and challenging orthopedic problem with poor self-healing ability and long treatment cycles. The difficult-to-heal bone defects cause a significant burden of medical expenses on patients. Currently, biomaterials with mechanical stability, long-lasting action, and osteogenic activity are considered as a suitable way to effectively heal bone defects. Here, an injectable double network (DN) hydrogel prepared using physical and chemical cross-linking methods is designed. The first rigid network is constructed using methylpropenylated hyaluronic acid (HAMA), while the addition of chitosan oligosaccharide (COS) forms a second flexible network by physical cross-linking. The mesoporous silica nanoparticles (MSN) loaded with bone morphogenetic protein-4 (BMP-4) were embedded into DN hydrogel, which not only enhanced the mechanical stability of the hydrogel, but also slowly released BMP-4 to achieve long-term skull repair. The designed composite hydrogel showed an excellent compression property and deformation resistance. In vitro studies confirmed that the HAMA/COS/MSN@BMP-4 hydrogel had good biocompatibility and showed great potential in supporting proliferation and osteogenic differentiation of mouse embryo osteoblast precursor (MC3T3-E1) cells. Furthermore, in vivo studies confirmed that the DN hydrogel successfully filled and closed irregular skull defect wounds, effectively promoted bone regeneration, and significantly promoted bone repair compared with the control group. In addition, HAMA/COS/MSN@BMP-4 hydrogel precursor solution can quickly form hydrogel in situ at the wound by ultraviolet light, which can be applied to the closure and repair of wounds of different shapes, which provides the new way for the treatment of bone defects.


Assuntos
Hidrogéis , Nanopartículas , Camundongos , Animais , Humanos , Hidrogéis/farmacologia , Hidrogéis/química , Osteogênese , Dióxido de Silício/farmacologia , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/farmacologia , Crânio/cirurgia , Crânio/lesões , Nanopartículas/química
8.
Molecules ; 29(6)2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38542880

RESUMO

Recombinant human bone morphogenetic protein 2 (rhBMP-2) is an FDA-approved growth factor for bone regeneration and repair in medical practice. The therapeutic effects of rhBMP-2 may be enhanced through specific binding to extracellular matrix (ECM)-like scaffolds. Here, we report the selection of a novel rhBMP-2-specific DNA aptamer, functionalization of the aptamer in an ECM-like scaffold, and its application in a cellular context. A DNA aptamer BA1 was evolved and shown to have high affinity and specificity to rhBMP-2. A molecular docking model demonstrated that BA1 was probably bound to rhBMP-2 at its heparin-binding domain, as verified with experimental competitive binding assays. The BA1 aptamer was used to functionalize a type I collagen scaffold, and fraction ratios were optimized to mimic the natural ECM. Studies in the myoblast cell model C2C12 showed that the aptamer-enhanced scaffold could specifically augment the osteo-inductive function of rhBMP-2 in vitro. This aptamer-functionalized scaffold may have value in enhancing rhBMP-2-mediated bone regeneration.


Assuntos
Aptâmeros de Nucleotídeos , Proteína Morfogenética Óssea 2 , Humanos , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 2/química , Aptâmeros de Nucleotídeos/farmacologia , Alicerces Teciduais/química , Simulação de Acoplamento Molecular , Regeneração Óssea , Fator de Crescimento Transformador beta/farmacologia , Proteínas Recombinantes/química
9.
Macromol Biosci ; 24(2): e2300245, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37572308

RESUMO

Microspheres (MSs) are ideal candidates as biological scaffolds loading with growth factors or cells for bone tissue engineering to repair irregular alveolar bone defects by minimally invasive injection. However, the high initial burst release of growth factor and low cell attachment limit the application of microspheres. The modification of microspheres often needs expensive experiments facility or complex chemical reactions, which is difficult to achieve and may bring other problems. In this study, a sol-grade nanoclay, laponite XLS is used to modify the surface of MSs to enhance its affinity to either positively or negatively charged proteins and cells without changing the interior structure of the MSs. Recombinant human bone morphogenetic protein-2 (rhBMP-2) is used as a representation of growth factor to check the osteoinduction ability of laponite XLS-modified MSs. By modification, the protein sustained release, cell loading, and osteoinduction ability of MSs are improved. Modified by 1% laponite XLS, the MSs can not only promote osteogenic differentiation of MC3T3-E1 cells by themselves, but also enhance the effect of the rhBMP-2 below the effective dose. Collectively, the study provides an easy and viable method to modify the biological behavior of microspheres for bone tissue regeneration.


Assuntos
Ácido Hialurônico , Osteogênese , Silicatos , Humanos , Ácido Hialurônico/farmacologia , Microesferas , Fator de Crescimento Transformador beta/farmacologia , Proteína Morfogenética Óssea 2/química , Regeneração Óssea , Proteínas Recombinantes/química
10.
Biochemistry (Mosc) ; 88(8): 1116-1125, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37758311

RESUMO

High efficiency of hybrid implants based on calcium-magnesium silicate ceramic, diopside, as a carrier of recombinant BMP-2 and xenogenic demineralized bone matrix (DBM) as a scaffold for bone tissue regeneration was demonstrated previously using the model of critical size cranial defects in mice. In order to investigate the possibility of using these implants for growing autologous bone tissue using in vivo bioreactor principle in the patient's own body, effectiveness of ectopic osteogenesis induced by them in intramuscular implantation in mice was studied. At the dose of 7 µg of BMP-2 per implant, dense agglomeration of cells, probably skeletal muscle satellite precursor cells, was observed one week after implantation with areas of intense chondrogenesis, initial stage of indirect osteogenesis, around the implants. After 12 weeks, a dense bone capsule of trabecular structure was formed covered with periosteum and mature bone marrow located in the spaces between the trabeculae. The capsule volume was about 8-10 times the volume of the original implant. There were practically no signs of inflammation and foreign body reaction. Microcomputed tomography data showed significant increase of the relative bone volume, number of trabeculae, and bone tissue density in the group of mice with BMP-2-containing implant in comparison with the group without BMP-2. Considering that DBM can be obtained in practically unlimited quantities with required size and shape, and that BMP-2 is obtained by synthesis in E. coli cells and is relatively inexpensive, further development of the in vivo bioreactor model based on the hybrid implants constructed from BMP-2, diopside, and xenogenic DBM seems promising.


Assuntos
Cálcio , Osteogênese , Camundongos , Humanos , Animais , Matriz Óssea , Microtomografia por Raio-X , Magnésio , Escherichia coli , Proteína Morfogenética Óssea 2/química , Silicatos de Magnésio/análise
11.
J Mater Sci Mater Med ; 34(8): 39, 2023 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-37498466

RESUMO

The aim of this study was to produce a composite of microporous ß-TCP filled with alginate-gelatin crosslinked hydrogel, clindamycin and bone morphogenetic protein (BMP-2) to prolong the drug-release behaviour for up to 28 days. The most promising alginate-di-aldehyde(ADA)-gelatin gel for drug release from microcapsules was used to fill microporous ß-TCP ceramics under directional flow in a special loading chamber. Dual release of clindamycin and BMP-2 was measured on days 1, 2, 3, 6, 9, 14, 21 and 28 by high performance liquid chromatography (HPLC) and enzyme-linked immunosorbent assay (ELISA). After release, the microbial efficacy of the clindamycin was checked and the biocompatibility of the composite was tested in cell culture. Clindamycin and the model substance FITC-protein A were released from microcapsules over 28 days. The clindamycin burst release was 43 ± 1%. For the loaded ceramics, a clindamycin release above the minimal inhibitory concentration (MIC) until day 9 and a burst release of 90.56 ± 2.96% were detected. BMP-2 was released from the loaded ceramics in low concentrations over 28 days. The release of active substances from ß-TCP and hydrogel have already been extensively studied. Directional flow loading is a special procedure in which the ceramic could act as a stabilizer in the bone and, as a biodegradable system, enables a single-stage surgical procedure. Whether ADA-gelatin gel is suitable for this procedure as a more biodegradable alternative to pure alginate or whether a dual release is possible in this composite has not yet been investigated.


Assuntos
Proteína Morfogenética Óssea 2 , Clindamicina , Alginatos/química , Proteína Morfogenética Óssea 2/química , Cápsulas , Cerâmica/química , Gelatina/química , Hidrogéis/química , Humanos , Animais
12.
Carbohydr Polym ; 315: 121002, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37230625

RESUMO

Bone morphogenetic protein 2 (BMP-2) is a potent osteoinductive factor that promotes bone formation. A major obstacle to the clinical application of BMP-2 is its inherent instability and complications caused by its rapid release from implants. Chitin based materials have excellent biocompatibility and mechanical properties, making them ideal for bone tissue engineering applications. In this study, a simple and easy method was developed to spontaneously form deacetylated ß-chitin (DAC-ß-chitin) gels at room temperature through a sequential deacetylation/self-gelation process. The structural transformation of ß-chitin to DAC-ß-chitin leads to the formation of self-gelling DAC-ß-chitin, from which hydrogels and scaffolds were prepared. Gelatin (GLT) accelerated the self-gelation of DAC-ß-chitin and increased the pore size and porosity of the DAC-ß-chitin scaffold. The DAC-ß-chitin scaffolds were then functionalized with a BMP-2-binding sulfate polysaccharide, fucoidan (FD). Compared with ß-chitin scaffolds, FD-functionalized DAC-ß-chitin scaffolds showed higher BMP-2 loading capacity and more sustainable release of BMP-2, and thus had better osteogenic activity for bone regeneration.


Assuntos
Proteína Morfogenética Óssea 2 , Hidrogéis , Proteína Morfogenética Óssea 2/química , Hidrogéis/química , Quitina , Preparações de Ação Retardada , Osteogênese , Regeneração Óssea , Engenharia Tecidual/métodos , Alicerces Teciduais/química
13.
Int J Biol Macromol ; 237: 124077, 2023 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-36934820

RESUMO

Covalent introduction of bioactive molecules is one of main strategies to significantly enhance the biological activities of bone repair materials. In this study, three most-commonly used chemical groups were respectively introduced on graphene (GP), followed by covalent binding with bone morphogenetic protein-2 (BMP-2) -derived peptides, ensuring that the same molar mass of peptides was bound to different functionalized GP (f-GP). Then the same amount of composites composed of different f-GP and peptides were respectively compounded with poly (lactic-co-glycolic acid) to fabricate 3D scaffolds. In vivo study demonstrated that the scaffolds containing ammonized GP covalently bound with the peptides through amide binding could reach best efficiency of promoting ectopic bone regeneration and repairing calvarial defect probably because the most positive charges on the peptide chain and surface of the ammonized GP could absorb more specific proteins in vivo and have better interactions with them, thereby differentiating most inducible cells into osteogenic cells. Our results indicate that the performances of scaffolds containing covalently bound bioactive molecules can be controlled by the covalent binding mode, and that our prepared scaffold containing ammonized GP covalently bound with the BMP-2-derived peptides through amide binding possess inspiring potential applicable prospects for bone tissue regeneration and engineering.


Assuntos
Grafite , Grafite/química , Osteogênese , Regeneração Óssea , Peptídeos/química , Proteína Morfogenética Óssea 2/química , Amidas , Alicerces Teciduais/química , Engenharia Tecidual/métodos
14.
Tissue Eng Regen Med ; 20(2): 155-156, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36964871

RESUMO

Bone morphogenic protein-2 (BMP-2)-conjugated three-dimensional (3-D)-printed poly (L-lactic acid)(PLLA) scaffold is likely promising as an effective bone substitute for enhancing bone regeneration of massive bone defects caused by tumor resection, traumatic injury, or congenital diseases. The authors developed a new bone substitute using a novel strategy composed of 3-D-printed PLLA scaffolds through a sequential coating of catechol-conjugated alginate (C-AL), BMP-2, and collagen (CO). The 3-D-printed PLLA scaffold was successfully obtained with 5 mm of diameter, 1 mm of thickness, 400 µm of pore size, 187-230 µm of grid thickness, and 82% of porosity. Alkaline phosphatase (ALP) activity of the BMP-2-immobilized PLLA scaffold in MC3T3-E1 and W-20-17 cells was more increased than BMP-2 itself due to the controlled release of BMP-2 from the scaffold. Tenfold new bone formation for the BMP-2-immobilized PLLA scaffold was obtained by micro-CT analysis than PLLA scaffold without BMP-2 weeks after 4 weeks of transplantation model mouse. Further another big animal model study should be performed before clinical trials.


Assuntos
Proteína Morfogenética Óssea 2 , Substitutos Ósseos , Alicerces Teciduais , Animais , Camundongos , Regeneração Óssea , Ácido Láctico , Engenharia Tecidual/métodos , Proteína Morfogenética Óssea 2/química
15.
Acta Biomater ; 162: 164-181, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36967054

RESUMO

Despite the glimmer of hope provided by the discovery and commercialization of bone morphogenetic protein-2 (BMP-2) as a bone graft substitute, side effects related to the use of supraphysiological doses have hindered its clinical usage. In this study, we compared the osteoinductive potential of BMP-2 homodimer with a heterodimer of BMP-2/7, both delivered via a collagen-hydroxyapatite (CHA) scaffold delivery system, with the aim to reduce the overall therapeutic BMP doses and the associated side-effects. We first show that the incorporation of hydroxyapatite in collagen-based BMP delivery systems is pivotal for achieving efficient BMP sequestration and controlled release. Using an ectopic implantation model, we then showed that the CHA+BMP-2/7 was more osteoinductive than CHA+BMP-2. Further evaluation of the molecular mechanisms responsible for this increased osteoinductivity at an early stage in the regeneration process indicated that the CHA+BMP-2/7 enhanced progenitor cell homing at the implantation site, upregulated the key transcriptomic determinants of bone formation, and increased the production of bone extracellular matrix components. Using fluorescently labelled BMP-2/7 and BMP-2, we demonstrated that the CHA scaffold provided a long-term delivery of both molecules for at least 20 days. Finally, using a rat femoral defect model, we showed that an ultra-low dose (0.5 µg) of BMP-2/7 accelerated fracture healing and performed at a level comparable to 20-times higher BMP-2 dose. Our results indicate that the sustained delivery of BMP-2/7 via a CHA scaffold could bring us a step closer in the quest for the use of physiological growth factor doses in fracture healing. STATEMENT OF SIGNIFICANCE: • Incorporation of hydroxyapatite (HA) in a collagen scaffold dramatically improves bone morphogenic protein (BMP) sequestration via biophysical interactions with BMP, thereby providing more controlled BMP release compared with pristine collagen. • We then investigate the molecular mechanisms responsible for increased osteoinductive potential of a heterodimer BMP-2/7 with is clinically used counterpart, the BMP-2 homodimer. • The superior osteoinductive properties of BMP-2/7 are a consequence of its direct positive effect on progenitor cell homing at the implantation site, which consequently leads to upregulation of cartilage and bone related genes and biochemical markers. • An ultra-low dose of BMP-2/7 delivered via a collagen-HA (CHA) scaffold leads to accelerated healing of a critical femoral defect in rats while a 20-times higher BMP-2 dose was required to achieve comparable results.


Assuntos
Substitutos Ósseos , Durapatita , Ratos , Animais , Durapatita/farmacologia , Colágeno/farmacologia , Colágeno/química , Osteogênese , Osso e Ossos , Consolidação da Fratura , Substitutos Ósseos/farmacologia , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 2/química , Regeneração Óssea
16.
Protein Expr Purif ; 206: 106245, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36805029

RESUMO

Human bone morphogenetic protein 2 (hBMP-2) plays a leading role in the process of osteogenesis and is one of the key components of osteoplastic materials, ensuring their high osteoinduction. In order to obtain a homodimeric form hBMP-2 using the E. coli expression system, a number of problems associated with refolding in vitro and purification from monomer and oligomeric forms must be solved. The developed method for co-expression of the target protein with chaperone proteins makes it possible to obtain the biologically active homodimeric form of hBMP-2 in vivo. Purification with simple ion-exchange sorbents without the use of denaturing reagents affecting the structure of the protein molecule provides a chromatographic purity of the product of at least 97%. The expressed hBMP-2 was identified by Western blotting and the LC-ESI-TOF mass spectrometry confirmed its molecular weight of 26052.72 Da. Circular dichroism spectroscopy showed that recombinant hBMP-2 has a native secondary structure.


Assuntos
Proteína Morfogenética Óssea 2 , Escherichia coli , Humanos , Proteína Morfogenética Óssea 2/química , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Recombinantes/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Osteogênese , Proteína Morfogenética Óssea 7/metabolismo
17.
Int J Biol Macromol ; 232: 123330, 2023 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-36681218

RESUMO

Bone regeneration is a complex process sequentially regulated by multiple cytokines at different stages. Vascular endothelial growth factor-A (VEGF-A) and bone morphogenetic protein-2 (BMP-2) are the two most important factors involved in this process, and the combination of the two can achieve better bone regeneration by coupling angiogenesis and osteogenesis. In this study, poly(lactic-co-glycolic acid) (PLGA) microspheres with core-shell structure (microcapsules) encapsulating VEGF-A or BMP-2 were prepared by coaxial channel injection and continuous fluid technology. The sequential release of two cytokines by microcapsules with different PLGA molecular weight and shell thickness and its performance in vitro were explored. It was demonstrated that the molecular weight of PLGA significantly affected the degradation and release kinetics of microcapsules, while the thickness of the shell can regulate the release in a finer level. VEGF-A encapsulated microcapsules with low molecular weight can induce vascular endothelial cells to form lumens structures in vitro at an early stage. And BMP-2 encapsulated microcapsules could promote osteogenic differentiation, but the effect could be delayed when the microcapsules were prepared with PLGA of 150 kDa. In conclusion, the core-shell PLGA microcapsules in this study can sequentially release VEGF-A and BMP-2 at different stages to simulate natural bone repair.


Assuntos
Osteogênese , Fator A de Crescimento do Endotélio Vascular , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Fator A de Crescimento do Endotélio Vascular/química , Ácido Poliglicólico/química , Ácido Láctico/química , Cápsulas , Células Endoteliais/metabolismo , Proteína Morfogenética Óssea 2/química , Citocinas , Alicerces Teciduais/química
18.
Int J Biol Macromol ; 227: 641-653, 2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36549614

RESUMO

A cavity defect inside the bone is formed by deformed cancellous bone from the fixation of the cortical bone, and consequently, abnormal bone healing occurs. Therefore, repairing cancellous bone defects is a remarkable topic in orthopedic surgery. In this study, we prepared bone marrow-derived stem cell (BMSC)-laden and bone morphogenetic protein-2 (BMP-2)-laden visible light-cured carboxymethyl chitosan (CMCS) hydrogels for cortical and cancellous bone healing. Proton nuclear magnetic resonance (1H NMR) analysis confirmed the methacrylation of CMCS (CMCSMA), resulting in 55 % of substitution. The higher concentration of CMCSMA hydrogel resulted in the lower swelling ratio, the larger viscosity, the slower degradation behavior, and the stronger compressive strength. The 5 w/v% hydrogel exhibited a controlled BMP-2 release for 14 days, while the 7 and 10 w/v% hydrogels displayed a controlled BMP-2 release for 28 days. Results of in vitro cytotoxicity and cell proliferation assays revealed the biocompatibility of the samples. In vivo animal tests demonstrated that BMSC- and BMP-2-laden 7 w/v% CMCSMA (CMCSMA+Cell+BMP-2) improved bone formation in the defected cortical and cancellous bones of the femur, as analyzed by micro-computed tomography (micro-CT) and histological evaluations. Consequently, we suggested that CMCSMA+Cell+BMP-2 can be a valuable scaffold for restoring cortical and cancellous bone defects.


Assuntos
Quitosana , Hidrogéis , Células-Tronco , Animais , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 2/química , Osso Esponjoso , Quitosana/química , Hidrogéis/farmacologia , Hidrogéis/química , Luz , Microtomografia por Raio-X , Medula Óssea
19.
Adv Healthc Mater ; 11(20): e2201339, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35941083

RESUMO

Cell-material interactions are regulated by mimicking bone extracellular matrix on the surface of biomaterials. In this regard, reproducing the extracellular conditions that promote integrin and growth factor (GF) signaling is a major goal to trigger bone regeneration. Thus, the use of synthetic osteogenic domains derived from bone morphogenetic protein 2 (BMP-2) is gaining increasing attention, as this strategy is devoid of the clinical risks associated with this molecule. In this work, the wrist and knuckle epitopes of BMP-2 are screened to identify peptides with potential osteogenic properties. The most active sequences (the DWIVA motif and its cyclic version) are combined with the cell adhesive RGD peptide (linear and cyclic variants), to produce tailor-made biomimetic peptides presenting the bioactive cues in a chemically and geometrically defined manner. Such multifunctional peptides are next used to functionalize titanium surfaces. Biological characterization with mesenchymal stem cells demonstrates the ability of the biointerfaces to synergistically enhance cell adhesion and osteogenic differentiation. Furthermore, in vivo studies in rat calvarial defects prove the capacity of the biomimetic coatings to improve new bone formation and reduce fibrous tissue thickness. These results highlight the potential of mimicking integrin-GF signaling with synthetic peptides, without the need for exogenous GFs.


Assuntos
Proteína Morfogenética Óssea 2 , Osteogênese , Ratos , Animais , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 2/química , Titânio , Diferenciação Celular , Matriz Extracelular , Regeneração Óssea , Peptídeos/farmacologia , Peptídeos/química , Materiais Biocompatíveis , Integrinas , Epitopos
20.
Biomed Res Int ; 2022: 4133562, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35342759

RESUMO

Mandibular bone regeneration is still a big challenge in those diabetic patients with poorly controlled blood glucose. In this study, we prepared a novel glucose-sensitive controlled-release fiber scaffold (PVA-HTCC/PEO-rhBMP2-glucose oxidase (PHPB-G)), which contained the recombinant human bone morphogenetic protein 2 (rhBMP2) by coaxial cospinning and grafted with glucose oxidase (GOD). We presented evidence that PHPB-G could undergo a series of structural changes with the blood glucose and promoted bone regeneration in diabetic rat. PHPB-G expanded the voids in nanofibers when blood glucose levels elevated. More importantly, its slow-release rhBMP2 effectively promoted the healing of bone defects. These data suggested that the PHPB-G delivery system may provide a potential treatment strategy for patients with severe diabetic alveolar bone defects.


Assuntos
Diabetes Mellitus , Osteogênese , Animais , Glicemia , Proteína Morfogenética Óssea 2/química , Proteína Morfogenética Óssea 2/farmacologia , Regeneração Óssea , Glucose/farmacologia , Glucose Oxidase/farmacologia , Humanos , Ratos , Proteínas Recombinantes/farmacologia , Alicerces Teciduais/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...