Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
BMC Oral Health ; 24(1): 69, 2024 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-38200461

RESUMO

OBJECTIVES: To evaluate the histological parameters and bone mechanical properties around implants with low primary stability (PS) in grafted bone substitutes within an oversized osteotomy. MATERIALS AND METHODS: An oversized osteotomy penetrating the double cortical bone layers was made on both femora of 24 New Zealand white rabbits. Bilaterally in the femur of all animals, 48 implants were installed, subdivided into four groups, corresponding to four prepared tissue-engineering bone complexes (TEBCs), which were placed between the implant surface and native bone wall: A: tricalcium phosphate ß (TCP-ß); B: autologous adipose derived-stem cells with TCP-ß (ASCs/TCP-ß); C: ASCs transfected with the enhanced-GFP gene with TCP-ß (EGFP-ASCs/TCP-ß); D: ASCs transfected with the BMP-2 gene with TCP-ß (BMP2-ASCs/TCP-ß). Trichrome fluorescent labeling was conducted. Animals were sacrificed after eight weeks. The trichromatic fluorescent labeling (%TFL), area of new bone (%NB), residual material (%RM), bone-implant contact (%BIC), and the removal torque force (RTF, N/cm) were assessed. RESULTS: ASCs were successfully isolated from adipose tissue, and the primary ASCs were induced into osteogenic, chondrogenic, and adipogenic differentiation. The BMP-2 overexpression of ASCs sustained for ten days and greatly enhanced the expression of osteopontin (OPN). At eight weeks post-implantation, increased %NB and RTF were found in all groups. The most significant value of %TFL, %BIC and lowest %RM was detected in group D. CONCLUSION: The low PS implants osseointegrate with considerable new bone in grafted TEBCs within an oversized osteotomy. Applying BMP-2 overexpressing ASCs-based TEBC promoted earlier osseointegration and more solid bone mechanical properties on low PS implants. Bone graft offers a wedging effect for the implant with low PS at placement and promotes osteogenesis on their surface in the healing period.


Assuntos
Substitutos Ósseos , Fosfatos de Cálcio , Implantes Dentários , Animais , Coelhos , Osseointegração , Osteotomia , Osteogênese , Corantes
2.
Int J Mol Sci ; 22(23)2021 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-34884885

RESUMO

With the limitation of autografts, the development of alternative treatments for bone diseases to alleviate autograft-related complications is highly demanded. In this study, a tissue-engineered bone was formed by culturing rat bone marrow cells (RBMCs) onto porous apatite-fiber scaffolds (AFSs) with three-dimensional (3D) interconnected pores using a radial-flow bioreactor (RFB). Using the optimized flow rate, the effect of different culturing periods on the development of tissue-engineered bone was investigated. The 3D cell culture using RFB was performed for 0, 1 or 2 weeks in a standard medium followed by 0, 1 or 2 weeks in a differentiation medium. Osteoblast differentiation in the tissue-engineered bone was examined by alkaline phosphatase (ALP) and osteocalcin (OC) assays. Furthermore, the tissue-engineered bone was histologically examined by hematoxylin and eosin and alizarin red S stains. We found that the ALP activity and OC content of calcified cells tended to increase with the culture period, and the differentiation of tissue-engineered bone could be controlled by varying the culture period. In addition, the employment of RFB and AFSs provided a favorable 3D environment for cell growth and differentiation. Overall, these results provide valuable insights into the design of tissue-engineered bone for clinical applications.


Assuntos
Células da Medula Óssea/fisiologia , Durapatita , Osteogênese , Engenharia Tecidual , Alicerces Teciduais , Fosfatase Alcalina/análise , Fosfatase Alcalina/metabolismo , Animais , Reatores Biológicos , Técnicas de Cultura de Células em Três Dimensões , Diferenciação Celular , Células Cultivadas , Ratos , Ratos Wistar , Células-Tronco/fisiologia
3.
J Musculoskelet Neuronal Interact ; 20(1): 142-148, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32131379

RESUMO

OBJECTIVE: to investigate the combined construction of injectable tissue-engineered bone with calcium phosphate bone cement composite (CPC) and bone marrow mesenchymal stem cells (BMMSCs). METHODS: The proliferation activity of BMMSCs encapsulated was detected by CCK8 method on the 7th day after its self-coagulation by CPC. qRT-PCR was used to detect the expressions of mRNA. The microcapsules of BMMSCs combined with CPC were completely filled in the defect site in the experimental group, and the control group not filled. The two groups were sutured and routinely reared, double upper limb X-ray examination performed after operation. RESULTS: Those of two groups were on the rise over time, which were higher at the 1st, 3rd, 5th and 7th days than those at the previous time points (all P<0.05). The relative expressions of ALP and CALCR at the 7th day were higher than those at the day in BMMSCs combined with the CPC group and BMMSCs group (all P<0.05). The relative expression of CALCR was significantly higher in BMMSCs combined with the CPC group than that in the BMMSCs group on the 7th day (P<0.05). CONCLUSION: With good cell activity and biological activity, the combined construction of the tissue-engineered bone with BMMSCs and CPC can be used as an ideal treatment material for bone tissue repair and connection.


Assuntos
Cimentos Ósseos/farmacologia , Transplante de Células-Tronco Mesenquimais/métodos , Rádio (Anatomia)/diagnóstico por imagem , Rádio (Anatomia)/lesões , Engenharia Tecidual/métodos , Animais , Fosfatos de Cálcio/farmacologia , Células Cultivadas , Terapia Combinada/métodos , Células-Tronco Mesenquimais/fisiologia , Coelhos , Rádio (Anatomia)/efeitos dos fármacos
4.
Cell Physiol Biochem ; 48(1): 361-370, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30016780

RESUMO

BACKGROUND/AIMS: Tissue engineering bone transplantation with bone marrow mesenchymal stem cells (BMSCs) is an effective technology to treat massive bone loss, while molecular regulation of the bone regeneration processes remains poorly understood. Here, we aimed to assess the role of interleukin-8 (IL-8) in the recruitment of host cells by seeded BMSCs and in the bone regeneration. METHODS: A transwell assay was performed to examine the role of IL-8/CXCR1/CXCR2/PI3k/Akt on the migration potential of hBMSCs. The in vitro chondrogenic differentiation of hBMSCs was assessed by examination of 2 chondrogenic markers, Sox9 and type 2 collagen (COL2). mBMSCs were used in tissue engineered bone (TEB) with/without IL-8 implanted into bone defect area with CXCR2 or Akt inhibitors. Density and Masson staining of the regenerated bone were assessed. The chondrogenesis was assessed by expression levels of associated proteins, Sox9 and COL2, by RT-qPCR and by immunohistochemistry. RESULTS: IL-8 may trigger in vitro migration of hBMSCs via CXCR2-mediated PI3k/Akt signaling pathway. IL-8 enhances osteogenesis in the TEB-implanted bone defect in mice. IL-8 induces chondrogenic differentiation of hBMSCs via CXCR2-mediated PI3k/Akt signaling pathway in vitro and in vivo. CONCLUSIONS: IL-8 enhances therapeutic effects of MSCs on bone regeneration via CXCR2-mediated PI3k/Akt signaling pathway.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Interleucina-8/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Interleucina-8B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Células da Medula Óssea/citologia , Osso e Ossos/patologia , Osso e Ossos/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Condrogênese/efeitos dos fármacos , Colágeno Tipo II/metabolismo , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Compostos de Fenilureia/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Receptores de Interleucina-8B/antagonistas & inibidores , Engenharia Tecidual
5.
Biochem Biophys Res Commun ; 495(1): 1264-1270, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29175390

RESUMO

To repair bone defects, we evaluate the in-vitro and in-vivo osteogenic activities of a novel tissue-engineered bone (TEB) by elaborately combining biomimetic calcium phosphate (BioCaP) granules with internally-incorporated simvastatin (SIM) and human adipose-derived stem cells (hASCs). First, we constructed BioCaP with SIM internally incorporated (SIM-BioCaP). Then we characterized the morphology and chemical composition of SIM-BioCaP. The release kinetics of SIM was monitored in vitro spectroscopically. Thereafter, we explored the in-vitro cellular responses of hASCs to SIM-BioCaP by performing scanning electron microscopy observation, proliferation assay, alkaline phosphatase (ALP) activity assay, alizarin red staining and real-time PCR. Finally, we investigated the in-vivo osteogenic activities of the novel TEB in a subcutaneous bone induction model in nude mice. We found that SIM was successfully incorporated internally in BioCaP and showed a slow release manner without significantly affecting the attachment and proliferation of hASCs. The released SIM from BioCaP could significantly enhance the proliferation, ALP activities, mineralized nodules formation and osteogenic genes of hASCs. The in-vivo tests showed this TEB could induce new bone formation while the other groups could not. Taken together, the present data show that this novel TEB represented a very promising construct to treat critical-volume bone defects.


Assuntos
Desenvolvimento Ósseo/fisiologia , Fosfatos de Cálcio/química , Transplante de Células-Tronco Mesenquimais/instrumentação , Osteogênese/fisiologia , Sinvastatina/administração & dosagem , Engenharia Tecidual/instrumentação , Alicerces Teciduais , Animais , Substitutos Ósseos , Células Cultivadas , Implantes de Medicamento/administração & dosagem , Implantes de Medicamento/química , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Nus , Osteoblastos/citologia , Osteoblastos/fisiologia , Engenharia Tecidual/métodos
6.
J Transl Med ; 16(1): 324, 2018 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-30470235

RESUMO

BACKGROUND: At present, many laboratories and hospitals all over the world are attempting and exploring the clinical transformation of this tissue engineered bone graft (TEBG) strategy. Many successful cases of bone tissue engineering (BTE) repair were based on young individuals. But there are little studies about the effectiveness of TEBG strategy in physiological aged individuals. METHODS: In this research, we studied whether aging factor has influence on the skull repair effect of Fetal-TEBG, at the level of the large animal models. We used the fetal bone marrow stromal cells (Fetal-BMSCs) as the seed cells, combining the decalcified bone matrix (DBM) scaffolds, to repair the skull defects of the aged goats and the young goats. The repair effects on both aged goat and young goat were compared by Micro-CT and histology examination. RESULTS: The skull defects of the young goats could be repaired better than that of the aged goats after 6 months by Fetal-TEBG; In the aged goats, although not completely repaired, the defects repaired by Fetal-TEBG was better than that repaired by the Control DBM scaffold. CONCLUSIONS: Aging factor has impact on the bone repair effect of Fetal-TEBG; and the BTE strategy is still efficacious even in the aged individuals. The improvement of the aged state may promote the repair effect of the BTE in the aged individuals.


Assuntos
Envelhecimento/fisiologia , Feto/citologia , Células-Tronco Mesenquimais/citologia , Crânio/fisiologia , Engenharia Tecidual/métodos , Animais , Diferenciação Celular , Proliferação de Células , Separação Celular , Forma Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Feminino , Cabras , Crânio/diagnóstico por imagem , Microtomografia por Raio-X
7.
Cell Tissue Res ; 370(1): 143-151, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28687929

RESUMO

The improved ectopic osteogenesis of cytotoxic T-lymphocyte-associated antigen 4-Ig-modified bone marrow mesenchymal stem cells (MSCs-CTLA4) has been demonstrated but the mechanisms involved remain to be determined. The extracellular matrix (ECM) has recently been reported to play a vital role in bone formation and periostin (POSTN) has been suggested as a key member in constructing the ECM in bone tissue. We found that POSTN expression in the MSCs-CTLA4 group is significantly enhanced compared with that in the MSCs group, not only in tissue-engineered bone (TEB) with femur heterotopic transplantation in vivo but also under the immune activation condition in vitro. This ectopic osteogenesis effect is in accordance with POSTN expression. We also found that the soluble POSTN treatment up-regulates osteogenic marker expression in MSCs, including runt-related transcription factor 2, collagen 1, osteocalcin, osterix, and alkaline phosphatase and calcium nodule formation. These effects are diminished when the soluble POSTN is neutralized. Our results demonstrate that POSTN promotes the osteogenic differentiation of MSCs and that CTLA4 enhances the ectopic osteogenesis of MSCs-CTLA4-based TEB, potentially by maintaining POSTN expression in xenotransplantation.


Assuntos
Antígeno CTLA-4/genética , Moléculas de Adesão Celular/genética , Células-Tronco Mesenquimais/citologia , Osteogênese , Adulto , Animais , Antígeno CTLA-4/metabolismo , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos BALB C , Engenharia Tecidual , Transplante Heterólogo , Regulação para Cima
8.
Clin Oral Investig ; 21(8): 2521-2534, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28101680

RESUMO

OBJECTIVES: The clinical standard for alveolar cleft osteoplasty is augmentation with autologous bone being available in limited amounts and might be associated with donor site morbidity. The aim of the present study was the creation of tissue-engineered bone grafts and their in vivo evaluation regarding their potential to promote osteogenesis in an alveolar cleft model. MATERIALS AND METHODS: Artificial bone defects with a diameter of 3.3 mm were created surgically in the palate of 84 adult Lewis rats. Four experimental groups (n = 21) were examined: bovine hydroxyl apatite/collagen (bHA) without cells, bHA with undifferentiated mesenchymal stromal cells (MSC), bHA with osteogenically differentiated MSC. In a control group, the defect remained empty. After 6, 9 and 12 weeks, the remaining defect volume was assessed by cone beam computed tomography. Histologically, the remaining defect width and percentage of bone formation was quantified. RESULTS: After 12 weeks, the remaining defect width was 60.1% for bHA, 74.7% for bHA with undifferentiated MSC and 81.8% for bHA with osteogenically differentiated MSC. For the control group, the remaining defect width measured 46.2% which was a statistically significant difference (p < 0.001). CONCLUSIONS: The study design was suitable to evaluate tissue-engineered bone grafts prior to a clinical application. In this experimental set-up with the described maxillary defect, no promoting influence on bone formation of bone grafts containing bHA could be confirmed. CLINICAL RELEVANCE: The creation of a sufficient tissue-engineered bone graft for alveolar cleft osteoplasty could preserve patients from donor site morbidity.


Assuntos
Enxerto de Osso Alveolar/métodos , Minerais/farmacologia , Osteogênese/fisiologia , Engenharia Tecidual/métodos , Animais , Diferenciação Celular , Células Cultivadas , Tomografia Computadorizada de Feixe Cônico , Fêmur/cirurgia , Masculino , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Modelos Animais , Ratos , Ratos Endogâmicos Lew , Retalhos Cirúrgicos
9.
J Surg Res ; 200(2): 544-51, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26521097

RESUMO

BACKGROUND: The pathological fracture is a most important complication during bone cyst and can be prevented by early focus clearance and bone grafting. Tissue-engineered bone (TEB) with outstanding osteogenesis is a better choice for bone repair. Here, we firstly reported that TEB was used to heal bone cyst. MATERIALS AND METHODS: The clinical data were collected from 23 patients who received bone defect repair separately with TEB or allogeneic bone (Allo-B) after erasion during 2004-2008. Allo-B had been as a control. The healing time and healing quality, the incidence of complications, the safety, and the bone grafting failure rate were compared. RESULTS: In TEB group, the follow-up time was 28 ± 15.48 months; nine cases were confirmed healed (3.45 ± 2.01 months), one case was cyst healing with defect, and one case had relapse. In Allo-B, 12 patients were followed up for 28.58 ± 20.44 months; seven cases were confirmed healed (6.75 ± 3.31 mo), four cases were cyst healing with defect, and one case had relapse. After operation, no statistically significant differences in bone healing and incidence of complications were observed between two groups, but the difference in bone healing time was statistically significant (P < 0.05). There was no else tumorigenesis in both groups. CONCLUSIONS: In treating simple bone cyst, Allo-B and TEB have considerable efficacy and safety; TEB is superior to Allo-B in respect of healing time; there is no rejection after TEB grafting but certain rejection after Allo-B grafting.


Assuntos
Cistos Ósseos/cirurgia , Transplante Ósseo/métodos , Calcâneo/cirurgia , Fêmur/cirurgia , Úmero/cirurgia , Rádio (Anatomia)/cirurgia , Engenharia Tecidual , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Seguimentos , Humanos , Masculino , Transplante Homólogo , Resultado do Tratamento , Adulto Jovem
10.
Adv Healthc Mater ; : e2401919, 2024 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-39155410

RESUMO

In this study, a new-generation tissue-engineered bone capable of temporally regulating the immune response, balancing proinflammatory and anti-inflammatory activities, and facilitating bone regeneration and repair to address the challenges of delayed healing and nonunion in large-sized bone defects, is innovatively developed. Using the innovative techniques including multiphysics-assisted combined decellularization, side-chain biochemical modification, and sterile freeze-drying, a novel photocurable extracellular matrix hydrogel, methacrylated bone-derived decellularized extracellular matrix (bdECM-MA), is synthesized. After incorporating the bdECM-MA with silicon-substituted calcium phosphate and bone marrow mesenchymal stem cells, the tissue-engineered bone is fabricated through digital light processing 3D bioprinting. This study provides in vitro confirmation that the engineered bone maintains high cellular viability while achieving MPa-level mechanical strength. Moreover, this engineered bone exhibits excellent osteogenesis, angiogenesis, and immunomodulatory functions. One of the molecular mechanisms of the immunomodulatory function involves the inhibition of the p38-MAPK pathway. A pioneering in vivo discovery is that the natural biomaterial-based tissue-engineered bone demonstrates sequential immunomodulatory properties that activate proinflammatory and anti-inflammatory responses in succession, significantly accelerating the repair of bone defects. This study provides a new research basis and an effective method for developing autogenous bone substitute materials and treating large-sized bone defects.

11.
Mater Today Bio ; 23: 100891, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38149016

RESUMO

The strategy of coupling the micro-vibration mechanical field with Ca/P ceramics to optimize the osteogenic microenvironment and enhance the functional activity of the cells can significantly improve the bone regeneration of the graft. However, the regulation mode and mechanism of this coupling strategy are not fully understood at present. This study investigated the influence of different waveforms of the electrical signals driving Microvibration Stimulation (MVS) on this coupling effect. The results showed that there were notable variances in calcium phosphate dissolution and redeposition, protein adsorption, phosphorylation of ERK1/2 and FAK signal pathways and activation of calcium channels such as TRPV1/Piezo1/Piezo2 in osteogenic microenvironment under the coupling action of hydroxyapatite (HA) ceramics and MVS driven by different electrical signal waveforms. Ultimately, these differences affected the osteogenic differentiation process of cells by a way of time-sequential regulation. Square wave-MVS coupled with HA ceramic can significantly delay the high expression time of characteristic genes (such as Runx2, Col-I and OCN) in MC3T3-E1 cells during in vitro the early, middle and late stage of differentiation, while maintain the high proliferative activity of MC3T3-E1 cells. Triangle wave signal-MVS coupled with HA ceramic promoted the osteogenic differentiation of cells in the early and late stages. Sine wave-MVS shows the effect on the process of osteogenic differentiation in the middle stage (such as the up-regulation of ALP synthesis and Col-I gene expression in the early stage of stimulation). In addition, Square wave-MVS showed the best coupling effect. The bone graft constructed under square wave-MVS formed new bone tissue and mature blood vessels only 2 weeks after subcutaneous implantation in nude mice. Our study provides a new non-invasive regulation model for precisely optimizing the osteogenic microenvironment, which can accelerate bone regeneration in bone grafts more safely, accurately and reliably.

12.
ACS Appl Mater Interfaces ; 15(1): 249-264, 2023 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-36548196

RESUMO

The regenerative repair of segmental bone defect (SBD) is an urgent problem in the field of orthopedics. Rapid induction of angiogenesis and osteoinductivity after implantation of scaffold is critical. In this study, a unique tissue engineering strategy with mixture of peripheral blood-derived mesenchymal stem cells (PBMSC) and endothelial progenitor cells (PBEPC) was applied in a 3D-printed biphasic calcium phosphate (BCP) scaffold with highly bioactive nano hydroxyapatite (nHA) coating (nHA/BCP) to construct a novel vascularized tissue engineered bone (VTEB) for rabbit femoral SBD repair. The 2D coculture of PBMSC and PBEPC showed that they could promote the osteogenic or angiogenic differentiation of the cells from each other, especially in the group of PBEPC/PBMSC = 75:25. Besides, the 3D coculture results exhibited that the nHA coating could further promote PBEPC/PBMSC adhesion, proliferation, and osteogenic and angiogenic differentiation on the BCP scaffold. In vivo experiments showed that among the four groups (BCP, BCP-PBEPC/PBMSC, nHA/BCP, and nHA/BCP-PBEPC/PBMSC), the nHA/BCP-PBEPC/PBMSC group induced the best formation of blood vessels and new bone and, thus, the good repair of SBD. It revealed the synergistic effect of nHA and PBEPC/PBMSC on the angiogenesis and osteogenesis of the BCP scaffold. Therefore, the construction of VTEB in this study could provide a possibility for the regenerative repair of SBD.


Assuntos
Engenharia Tecidual , Alicerces Teciduais , Animais , Coelhos , Engenharia Tecidual/métodos , Hidroxiapatitas/farmacologia , Durapatita/farmacologia , Osteogênese , Diferenciação Celular , Regeneração Óssea
13.
J Food Biochem ; 46(10): e14329, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35867029

RESUMO

During the implantation of functional tissue-engineered constructs for treating bone defects, a functional vascular network is critical for the survival of the construct. One strategy to achieve rapid angiogenesis for this application is the co-culture of outgrowth endothelial cells (OECs) and primary human osteoblasts (POBs) within a scaffold prior to implantation. In the present study, we aim to investigate whether Astragalus polysaccharide (APS) promotes angiogenesis or vascularization via the TLR4 signaling pathway in a co-culture of OECs and POBs. The co-cultures were treated with various concentrations of APS for 24 h and, subsequently, another 7 days, followed by CD31 staining and analysis of micro-vessel-formation areas using software. Additionally, APS (0.4 mg/ml for 24 h) was added to monocultures of OECs or POBs for evaluating proliferation, apoptosis, angiogenesis, osteogenesis, TLR4 signaling pathway, and inflammatory cytokine release. We found that APS promoted angiogenesis in the co-culture at the optimal concentration of 0.4 mg/ml. TLR4 activation by APS up-regulated the expression level of TLR4/MyD88 and enhanced angiogenesis and osteogenesis in monocultures of OECs and POBs. The levels of E-selectin adhesion molecules, three cytokines (IL-6, TNF-α, and IFN-γ), and VEGF and PDGF-BB, which can induce angiogenesis, increased significantly (p < .05) following APS treatment. Therefore, APS appears to promote angiogenesis and ossification in the co-culture system via the TLR4 signaling pathway. PRACTICAL APPLICATIONS: This study demonstrates that APS may promote angiogenesis and osteocyte proliferation in OEC and POB co-culture systems through the MyD88-dependent TLR4 signaling pathway. APS might represent a potential therapeutic strategy in tissue-engineered bone implantation for the treatment of large bone defects; additionally, it has the advantage of safety, as it exhibits low or no side effects. In the future, it is expected to be used in vitro for the construction of tissue-engineered bone and in vivo after implantation in patients with bone defects for promoting rapid vascularization and ossification of tissue-engineered bone and early fusion with the recipient's bone. In addition, as a food additive, Astragalus membranaceus can be used as a tonic material in patients recovering from a fracture for promoting blood-vessel formation at the fracture site and fracture recovery. Combining traditional Chinese medicine with tissue engineering can provide further strategies for promoting the development of regenerative medicine.


Assuntos
Células Endoteliais , Receptor 4 Toll-Like , Becaplermina/metabolismo , Selectina E/metabolismo , Células Endoteliais/metabolismo , Aditivos Alimentares , Humanos , Interleucina-6/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Neovascularização Fisiológica , Polissacarídeos/metabolismo , Polissacarídeos/farmacologia , Transdução de Sinais , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Materials (Basel) ; 15(23)2022 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-36499970

RESUMO

Bone defects caused by various factors may cause morphological and functional disorders that can seriously affect patient's quality of life. Autologous bone grafting is morbid, involves numerous complications, and provides limited volume at donor site. Hence, tissue-engineered bone is a better alternative for repair of bone defects and for promoting a patient's functional recovery. Besides good biocompatibility, scaffolding materials represented by hydroxyapatite (HA) composites in tissue-engineered bone also have strong ability to guide bone regeneration. The development of manufacturing technology and advances in material science have made HA composite scaffolding more closely related to the composition and mechanical properties of natural bone. The surface morphology and pore diameter of the scaffold material are more important for cell proliferation, differentiation, and nutrient exchange. The degradation rate of the composite scaffold should match the rate of osteogenesis, and the loading of cells/cytokine is beneficial to promote the formation of new bone. In conclusion, there is no doubt that a breakthrough has been made in composition, mechanical properties, and degradation of HA composites. Biomimetic tissue-engineered bone based on vascularization and innervation show a promising future.

15.
Int J Surg Case Rep ; 99: 107569, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36108376

RESUMO

INTRODUCTION AND IMPORTANCE: We used induced membrane combined with tissue-engineered bone (TEB) to repair the 14-cm juvenile ulnar defect formed after osteomyelitis debridement. The TEB was completely transformed into autologous bone after 4-year follow-up. CASE PRESENTATION: A 13-year-old male was hospitalized because of right ulna chronic osteomyelitis. After focal debridement, the total length of ular defect was 14 cm. Anti-infective bone cement was filled in the bone defect area. ß-Tricalcium phosphate (ß-TCP) was used as TEB scaffold. Autologous iliac bone marrow stromal cells (BMSCs) were cultured in vitro and were planted on ß-TCP scaffold to form TEB 3 weeks later. 47 months after implantation of TEB, the repaired ulna had continuous and smooth bone cortex, completely ossification of TEB, completely recanalization of medullary cavity. The upper limb function DASH score was 35. CLINICAL DISCUSSION: Masquelet put forward the concept of "induced membrane" and applied this technique on bone defects treatment formed after debridement of osteomyelitis. ß-Tricalcium phosphate (ß-TCP) is artificial bone materials commonly used in clinical. In this case, the seed cells used were autologous BMSCs and the culture medium was autologous serum. Cytokines promoting cell growth and differentiation were not used. CONCLUSION: The results of this case showed that TEB combined with induced membrane could repair ulna segmental bone defects as long as 14 cm in adolescents. This technique gives one alternative method to repair juvenile bone defects caused by osteomyelities of trauma. More clinical cases are needed to verify the effectiveness of this technique in the next.

16.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 36(5): 625-632, 2022 May 15.
Artigo em Chinês | MEDLINE | ID: mdl-35570639

RESUMO

Objective: To evaluate the biological effect on vascularization during bone repair of prevascularized porous ß-tricalcium phosphate (ß-TCP) tissue engineered bone (hereinafter referred to as prevascularized tissue engineered bone), which was established by co-culture of endothelial progenitor cells (EPCs) and bone marrow mesenchymal stem cells (BMSCs) based on tissue engineering technology. Methods: EPCs and BMSCs were isolated from iliac bone marrow of New Zealand white rabbits by density gradient centrifugation and differential adhesion method. The cells were identified by immunophenotypic detection, BMSCs-induced differentiation, and EPCs phagocytosis. After identification, the third-generation cells were selected for subsequent experiments. First, in vitro tubule formation in EPCs/BMSCs direct contact co-culture (EPCs/BMSCs group) was detected by Matrigel tubule formation assay and single EPCs (EPCs group) as control. Then, the prevascularized tissue engineered bone were established by co-culture of EPCs/BMSCs in porous ß-TCP scaffolds for 7 days (EPCs/BMSCs group), taking EPCs in porous ß-TCP scaffolds as a control (EPCs group). Scanning electron microscopy and laser scanning confocal microscopy were used to observe the adhesion, proliferation, and tube formation of cells. Femoral condyle defect models of 12 New Zealand white rabbits were used for implantation of prevascularized tissue engineered bone as the experimental group ( n=6) and porous ß-TCP scaffold as the control group ( n=6). The process of vascularization of ß-TCP scaffolds were observed. The numbers, diameter, and area fraction of neovascularization were quantitatively evaluated by Microfill perfusion, Micro-CT scanning, and vascular imaging under fluorescence at 4 and 8 weeks. Results: The isolated cells were BMSCs and EPCs through identification. EPCs/BMSCs co-culture gradually formed tubular structure. The number of tubules and branches, and the total length of tubules formed in the EPCs/BMSCs group were significantly more than those in the EPCs group on Matrigel ( P<0.05) after 6 hours. After implanting and culturing in porous ß-TCP scaffold for 7 days, EPCs formed cell membrane structure and attached to the material in EPCs group, and the cells attached more tightly, cell layers were thicker, the number of cells and the formation of tubular structures were significantly more in the EPCs/BMSCs group than in the EPCs group. At 4 weeks after implantation, neovascularization was observed in both groups. At 8 weeks, remodeling of neovascularization occurred in both groups. The number, diameter, and area fraction of neovascularization in the experimental group were higher than those in the control group ( P<0.05), except for area fraction at 4 weeks after implantation ( P>0.05). Conclusion: The prevascularized tissue engineered bone based on direct contact co-culture of BMSCs and EPCs can significantly promote the early vascularization process during bone defects repair.


Assuntos
Engenharia Tecidual , Alicerces Teciduais , Animais , Células da Medula Óssea , Osso e Ossos , Fosfatos de Cálcio/farmacologia , Células Cultivadas , Osteogênese , Porosidade , Coelhos , Engenharia Tecidual/métodos , Alicerces Teciduais/química
17.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 36(1): 102-110, 2022 Jan 15.
Artigo em Chinês | MEDLINE | ID: mdl-35038807

RESUMO

OBJECTIVE: To investigate the role and regulatory mechanism of ring finger protein 11 (RNF11) on Akt signaling pathway in the process of osteogenesis of bone marrow mesenchymal stem cells (BMSCs) to provide ideas for further clarifying its osteogenesis mechanism and its use in clinical treatment in the future. METHODS: BMSCs were isolated and cultured from fresh bone marrow of healthy donors and subcultured. The 4th generation cells were used in experiments after identification by flow cytometry, and osteogenic, chondrogenic, and adipogenic induction. BMSCs were cultured in osteogenic differentiation medium for 0-14 days. The degree of osteogenic differentiation was detected by Alizarin red staining and alkaline phosphatase (ALP) staining, and the protein expression of RNF11 was detected by Western blot. The 4th generation BMSCs were divided into blank control group (group A), empty lentivirus (Lv-NC) group (group B), and knockdown RNF11 (Lv-ShRNF11) group (group C). Osteogenesis was induced and cultured for 0-14 days. The expression of RNF11 protein was detected by Western blot, the degree of osteogenic differentiation was detected by Alizarin red staining and ALP staining, and the relative mRNA expressions of Runx2, osteocalcin (OCN), and osteopontin (OPN) were detected by real-time fluorescence quantitative PCR (qRT-PCR). The protein relative expressions of Akt, Smad1/5/8, and ß-catenin signaling pathway were detected by Western blot, expressed as the ratio before and after phosphorylation. In order to study the effect mechanism of RNF11 on Akt signaling pathway, the 4th generation BMSCs were divided into Lv-NC transfection group (group A1), Lv-ShRNF11 transfection group (group B1), and Lv-ShRNF11 transfection supplemented with Akt signaling pathway activator SC79 group (group C1). The protein relative expressions of RNF11 and Akt signaling pathway were detected by Western blot, the related osteogenesis indexes were detected by Alizarin red staining, ALP staining, and qRT-PCR. RESULTS: The flow cytometry, and osteogenic, chondrogenic, adipogenic induction culture identification showed that the isolated and cultured cells were BMSCs. The protein relative expression of RNF11 increased gradually with the extension of osteogenic differentiation time ( P<0.05); after knockdown RNF11, Alizarin red and ALP stainings showed that the degree of osteogenic differentiation of BMSCs in group C were significantly lower than those in groups A and B, and qRT-PCR detection showed that the relative expression of Runx2, OCN, and OPN mRNA significantly decreased ( P<0.05). The protein relative expressions of RNF11 and Akt signaling pathway significantly increased with the extensions of osteogenic differentiation time ( P<0.05). After knockdown RNF11, the protein relative expression of Akt signaling pathway in group C was significantly lower than that in groups A and B ( P<0.05), while Smad1/5/8 and ß-catenin signaling pathway had no significant effect ( P>0.05). Compared with group A1, the protein relative expression of RNF11 in groups B1 and C1 significantly decreased ( P<0.05). Compared with groups A1 and C1, the protein relative expression of Akt signaling pathway in group B1 was significantly lower ( P<0.05); Alizarin red and ALP stainings showed that the degree of osteogenic differentiation of BMSCs in group C1 were slightly lower than that of group A1 ( P>0.05), but significantly higher than that of group B1 ( P<0.05); qRT-PCR detection showed that the relative expressions of Runx2, OCN, and OPN mRNA in group C1 were slightly lower than those of group A1 ( P>0.05), but were significantly higher than those of group B1 ( P<0.05). CONCLUSION: RNF11 promotes the differentiation of BMSCs into osteoblasts by positively regulating the activation level of Akt signaling pathway. RNF11 can be used as a potential target to improve the bone repair efficacy of BMSCs and treat bone metabolic diseases.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Células da Medula Óssea , Diferenciação Celular , Células Cultivadas , Proteínas de Ligação a DNA , Humanos , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais
18.
Mater Today Bio ; 16: 100382, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36033373

RESUMO

Large bone defects remain an unsolved clinical challenge because of the lack of effective vascularization in newly formed bone tissue. 3D bioprinting is a fabrication technology with the potential to create vascularized bone grafts with biological activity for repairing bone defects. In this study, vascular endothelial cells laden with thermosensitive bio-ink were bioprinted in situ on the inner surfaces of interconnected tubular channels of bone mesenchymal stem cell-laden 3D-bioprinted scaffolds. Endothelial cells exhibited a more uniform distribution and greater seeding efficiency throughout the channels. In vitro, the in situ bioprinted endothelial cells can form a vascular network through proliferation and migration. The in situ vascularized tissue-engineered bone also resulted in a coupling effect between angiogenesis and osteogenesis. Moreover, RNA sequencing analysis revealed that the expression of genes related to osteogenesis and angiogenesis is upregulated in biological processes. The in vivo 3D-bioprinted in situ vascularized scaffolds exhibited excellent performance in promoting new bone formation in rat calvarial critical-sized defect models. Consequently, in situ vascularized tissue-engineered bones constructed using 3D bioprinting technology have a potential of being used as bone grafts for repairing large bone defects, with a possible clinical application in the future.

19.
Materials (Basel) ; 14(4)2021 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-33572321

RESUMO

Today, materials designed for bone regeneration are requested to be degradable and resorbable, bioactive, porous, and osteoconductive, as well as to be an active player in the bone-remodeling process. Multiphasic silica/collagen Xerogels were shown, earlier, to meet these requirements. The aim of the present study was to use these excellent material properties of silica/collagen Xerogels and to process them by additive manufacturing, in this case 3D plotting, to generate implants matching patient specific shapes of fractures or lesions. The concept is to have Xerogel granules as active major components embedded, to a large proportion, in a matrix that binds the granules in the scaffold. By using viscoelastic alginate as matrix, pastes of Xerogel granules were processed via 3D plotting. Moreover, alginate concentration was shown to be the key to a high content of irregularly shaped Xerogel granules embedded in a minimum of matrix phase. Both the alginate matrix and Xerogel granules were also shown to influence viscoelastic behavior of the paste, as well as the dimensionally stability of the scaffolds. In conclusion, 3D plotting of Xerogel granules was successfully established by using viscoelastic properties of alginate as matrix phase.

20.
Front Immunol ; 12: 689269, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34248977

RESUMO

Bone defects are a common orthopaedic concern, and an increasing number of tissue-engineered bones (TEBs) are used to repair bone defects. Allogeneic mesenchymal stem cells (allo-MSCs) are used as seed cells in many approaches to develop TEB constructs, but the immune response caused by allogeneic transplantation may lead to transplant failure. V gamma 4 T (Vγ4T) cells play an important role in mediating the immune response in the early stage after transplantation; therefore, we wanted to verify whether suppressing Vγ4T cells by herpesvirus entry mediator (HVEM)/B and T lymphocyte attenuator (BTLA) signalling can promote MSCs osteogenesis in the transplanted area. In vitro experiments showed that the osteogenic differentiation of MSCs and Vγ4T cells was weakened after co-culture, and an increase in interleukin-17 (IL-17) and interferon-γ (IFN-γ) levels was detected in the culture supernatant. HVEM-transfected MSCs (MSCs-HVEM) still exhibited osteogenic differentiation activity after co-culture with Vγ4T cells, and the levels of IL-17 and IFN-γ in the co-culture supernatant were significantly reduced. In vivo experiments revealed that inflammation in the transplanted area was reduced and osteogenic repair was enhanced after Vγ4T cells were removed. MSCs-HVEM can also consistently contribute to reduced inflammation in the transplanted area and enhanced bone repair in wild-type (WT) mice. Therefore, our experiments verified that HVEM can promote the osteogenesis of allo-MSCs by inhibiting IL-17 and IFN-γ secretion from Vγ4T cells.


Assuntos
Interferon gama/imunologia , Interleucina-17/imunologia , Células-Tronco Mesenquimais/citologia , Osteogênese , Membro 14 de Receptores do Fator de Necrose Tumoral/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Células Cultivadas , Técnicas de Cocultura , Diagnóstico por Imagem , Feminino , Fêmur/diagnóstico por imagem , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores Imunológicos/imunologia , Engenharia Tecidual
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa