Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
J Orthop Translat ; 33: 174-185, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35495963

RESUMO

Objective: To prepare adipose-derived stem cells (ADSCs)-embedded alginate-gelatinemicrospheres (Alg-Gel-ADSCs MSs) by electrospray and evaluate their feasibility for cartilage tissue engineering. To observe the efficacy of Alg-Gel-ADSCs MSs in repairing articular cartilage defects in SD rats. Methods: ADSCs were isolated and characterized by performing induced differentiation and flow cytometry assays. Alginate-gelatine microspheres with different gelatine concentrations were manufactured by electrospraying, and the appropriate alginate-gelatine concentration and ratio were determined by evaluating microsphere formation. Alg-Gel-ADSCs MSs were compared with Alg-ADSCs MSs through the induction of chondrogenic differentiation and culture. Their feasibility for cartilage tissue engineering was analysed by performing Live/Dead staining, cell proliferation analysis, toluidine blue staining and a glycosaminoglycan (GAG) content analysis. Alg-Gel-ADSCs MSs were implanted in the cartilage defects of SD rats, and the cartilage repair effect was evaluated at different time points. The evaluation included gross observations and histological evaluations, fluorescence imaging tracking, immunohistochemical staining, microcomputed tomography (micro-CT) and a CatWalk evaluation. Results: The isolated ADSCs showed multidirectional differentiation and were used for cartilage tissue engineering. Using 1.5 w:v% alginate and 0.5 w:v% gelatine (Type B), we successfully prepared nearly spherical microspheres. Compared with alginate microspheres, alginate gel increased the viability of ADSCs and promoted the proliferation and chondrogenesis of ADSCs. In our experiments on knee cartilage defects in SD rats in vivo, the Alg-Gel-ADSCs MSs showed superior cartilage repair in cell resides, histology evaluation, micro-CT imaging and gait analysis. Conclusions: Microspheres composed of 1.5 w:v% alginate-0.5 w:v% gelatine increase the viability of ADSCs and supported their proliferation and deposition of cartilage matrix components. ADSCs embedded in 1.5 w:v% alginate-0.5 w:v% gelatine microspheres show superior repair efficacy and prospective applications in cartilage tissue repair. The translational potential of this article: In this study, injectable adipose-derived stem cells-embedded alginate-gelatin microspheres (Alg-Gel-ADSCs MSs) were prepared by the electrospray . Compared with the traditional alginate microspheres, its support ability for ADSCs is better and shows a better repair effect. This study provides a promising strategy for cartilage tissue regeneration.

2.
J Orthop Translat ; 27: 132-138, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33786320

RESUMO

BACKGROUND: Osteonecrosis of the femoral head (ONFH) is a refractory disease due to its unclear pathomechanism. Neither conservative treatment nor surgical treatment during the early stage of ONFH achieves satisfactory results. Therefore, this study aims to explore the available evidence on the effect of zoledronic acid on early-stage ONFH. METHODS: For groups were established:the Normal group, model group, Normal saline group(NS group) and zoledronic acid-treated group. The blood supply to the femoral head of animals in the model group and zoledronic acid-treated group was interrupted via a surgical procedure, and zoledronic acid was then locally administered to the femoral head. Four weeks after surgery, all the hips were harvested and evaluated by micro-CT and histopathology(H&E staining, TRAP staining, Toluidine blue staining and masson staining). RESULTS: The values of BMD, BS/BV and Tb.Th in the Normal group and zoledronic acid-treated group were significantly higher than those in the model group and NS group (p â€‹< â€‹0.05). The outcome of H&E staining, Toluidine blue staining and masson staining were consistent with that of micro-CT. CONCLUSION: The local administration of zoledronic acid in the femoral head had positive effects on the bone structure of the femoral head in a modified rat model of traumatic ONFH and offered a promising therapeutic strategy during the early stage of ONFH. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: This article could provide a choice for treating patients who have osteonecrosis of femora head and can be the basic research for advanced development over this disease.

3.
J Orthop Translat ; 27: 101-109, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33520655

RESUMO

Articular cartilage regeneration is one of the challenges faced by orthopedic surgeons. Microcarrier applications have made great advances in cartilage tissue engineering in recent years and enable cost-effective cell expansion, thus providing permissive microenvironments for cells. In addition, microcarriers can be loaded with proteins, factors, and drugs for cartilage regeneration. Some microcarriers also have the advantages of injectability and targeted delivery. The application of microcarriers with these characteristics can overcome the limitations of traditional methods and provide additional advantages. In terms of the transformation potential, microcarriers have not only many advantages, such as providing sufficient and beneficial cells, factors, drugs, and microenvironments for cartilage regeneration, but also many application characteristics; for example, they can be injected to reduce invasiveness, transplanted after microtissue formation to increase efficiency, or combined with other stents to improve mechanical properties. Therefore, this technology has enormous potential for clinical transformation. In this review, we focus on recent advances in microcarriers for cartilage regeneration. We compare the characteristics of microcarriers with other methods for repairing cartilage defects, provide an overview of the advantages of microcarriers, discuss the potential of microcarrier systems, and present an outlook for future development. TRANSLATIONAL POTENTIAL OF THIS ARTICLE: We reviewed the advantages and recent advances of microcarriers for cartilage regeneration. This review could give many scholars a better understanding of microcarriers, which can provide doctors with potential methods for treating patients with cartilage injure.

4.
Orthop Surg ; 12(6): 1913-1922, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33185022

RESUMO

OBJECTIVE: The aim of the present paper was to evaluate the results of one-stage total hip arthroplasty (THA) for patients with bilateral Crowe type IV developmental dysplasia of the hip (DDH). METHODS: Data for 58 patients (116 hips) with bilateral Crowe type IV DDH who had one-stage THA performed by the same surgeon during the period of April 2008 to February 2019 were retrospectively reviewed. The mean age of the patients was 37.3 years; 5 were men and 53 were women. All patients underwent THA through the posterolateral approach using the Pinnacle acetabular cup, a ceramic-on-ceramic bearing, and the modular S-ROM stem. Subtrochanteric shortening osteotomy was performed on 86/116 hips. Intraoperative conditions were recorded. Radiographic and functional outcomes were evaluated, and complications were recorded. RESULTS: All patients were followed up for an average of 71.3 ± 37.6 months (range, 12-140). The mean operative time was 276.5 ± 57.9 min (range, 175-540). The mean intraoperative blood loss was 933.6 ± 400.8 mL (range, 300-2000). The mean transfusion requirement was 1778 ± 798.0 mL (range, 575-4550). The mean length of hospital stay was 8.6 ± 3.7 days (range, 5-22). At the final follow-up, no loosening of acetabular and femoral components was observed. No osteolysis and heterotopic ossification occurred. The mean Harris hip scores were improved from 55.4 ± 14.3 preoperatively to 91.3 ± 4.2 postoperatively (P < 0.001) In terms of complications, no perioperative deaths were recorded. Deep vein thrombosis occurred in 1 hip, with no pulmonary embolism. Intraoperative femur fracture occurred in 3 hips, nerve injury in 1 hip, and leg length discrepancy in 1 patient. Postoperative dislocation occurred in 5 hips and nonunion in 1 hip. CONCLUSION: Our data demonstrated that one-stage bilateral THA for bilateral Crowe type IV DDH is feasible and can effectively restore hip function.


Assuntos
Artroplastia de Quadril/métodos , Luxação Congênita de Quadril/cirurgia , Prótese de Quadril , Complicações Pós-Operatórias/etiologia , Desenho de Prótese , Adolescente , Adulto , Idoso , Artroplastia de Quadril/instrumentação , Avaliação da Deficiência , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Adulto Jovem
5.
Int J Nanomedicine ; 15: 7155-7171, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33061373

RESUMO

OBJECTIVE: Tissue engineering approaches seem to be an attractive therapy for tendon rupture. Novel injectable porous gelatin microcryogels (GMs) can promote cell attachment and proliferation, thus facilitating the repair potential for target tissue regeneration. The research objectives of this study were to assess the efficacy of tissue-like microunits constructed by multiple GMs laden with adipose-derived mesenchymal stem cells (ASCs) in accelerated tendon regeneration in a rat model. METHODS: Through a series of experiments, such as isolation and identification of ASCs, scanning electron microscopy, mercury intrusion porosimetry (MIP), laser scanning confocal microscopy and the CCK-8 test, the biocompatibility of GMs was evaluated. In an in vivo study, 64 rat right transected Achilles tendons were randomly divided into four groups: the ASCs+GMs group (microunits aggregated by multiple ASC-laden GMs injected into the gap), the ASCs group (ASCs injected into the gap), the GMs group (GMs injected into the gap) and the blank defect group (non-treated). At 2 and 4 weeks postoperatively, the healing tissue was harvested to evaluate the gross observation and scoring, biomechanical testing, histological staining and quantitative scoring. Gait analysis was performed over time. The 64 rats were randomly assigned into 4 groups: (1) micro-unit group (ASCs+GMs) containing ASC (105)-loaded 120 GMs in 60 µL DMEM; (2) cell control group (ASCs) containing 106 ASCs in 60 µL DMEM; (3) GM control group (GMs) containing 120 blank GMs in 60 µL DMEM; (4) blank defect group (Defect) containing 60 µL DMEM, which were injected into the defect sites. All animals were sacrificed at 2 and 4 weeks postsurgery (Table 1). RESULTS: In an in vitro study, GMs (from 126 µm to 348 µm) showed good porosities and a three-dimensional void structure with a good interpore connectivity of the micropores and exhibited excellent biocompatibility with ASCs. As the culture time elapsed, the extracellular matrix (ECM) secreted by ASCs encased the GMs, bound multiple microspheres together, and then formed active tendon tissue-engineering microunits. In animal experiments, the ASCs+GMs group and the ASCs group showed stimulatory effects on Achilles tendon healing. Moreover, the ASCs+GMs group was the best at improving the macroscopic appearance, histological morphology, Achilles functional index (AFI), and biomechanical properties of repair tissue without causing adverse immune reactions. CONCLUSION: Porous GMs were conducive to promoting cell proliferation and facilitating ECM secretion. The ASCs-GMs matrices showed an obvious therapeutic efficiency for Achilles tendon rupture in rats.


Assuntos
Tendão do Calcâneo/patologia , Tecido Adiposo/citologia , Criogéis/farmacologia , Células-Tronco Mesenquimais/citologia , Traumatismos dos Tendões/patologia , Traumatismos dos Tendões/terapia , Cicatrização/efeitos dos fármacos , Doença Aguda , Animais , Materiais Biocompatíveis/química , Fenômenos Biomecânicos , Diferenciação Celular , Modelos Animais de Doenças , Fluorescência , Gelatina/química , Masculino , Fenótipo , Porosidade , Ratos Sprague-Dawley , Ruptura , Engenharia Tecidual
6.
Acta Biomater ; 114: 31-52, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32652223

RESUMO

In the absence of timely and proper treatments, injuries to articular cartilage (AC) can lead to cartilage degeneration and ultimately result in osteoarthritis. Regenerative medicine and tissue engineering techniques are emerging as promising approaches for AC regeneration and repair. Although the use of cell-seeded scaffolds prior to implantation can regenerate and repair cartilage lesions to some extent, these approaches are still restricted by limited cell sources, excessive costs, risks of disease transmission and complex manufacturing practices. Recently developed acellular scaffold approaches that rely on the recruitment of endogenous cells to the injured sites avoid these drawbacks and offer great promise for in situ AC regeneration. Multiple endogenous stem/progenitor cells (ESPCs) are found in joint-resident niches and have the capability to migrate to sites of injury to participate in AC regeneration. However, the natural recruitment of ESPCs is insufficient, and the local microenvironment is hostile after injury. Hence, an endogenous cell recruitment strategy based on the combination of chemoattractants and acellular scaffolds to effectively and specifically recruit ESPCs and improve local microenvironment may provide new insights into in situ AC regeneration. This review provides a brief overview of: (1) the status of endogenous cell recruitment strategy; (2) the subpopulations, potential migration routes (PMRs) of joint-resident ESPCs and their immunomodulatory and reparative effects; (3) chemoattractants and their potential adverse effects; (4) scaffold-based drug delivery systems (SDDSs) that are utilized for in situ AC regeneration; and (5) the challenges and future perspectives of endogenous cell recruitment strategy for AC regeneration. STATEMENT OF SIGNIFICANCE: Although the endogenous cell recruitment strategy for articular cartilage (AC) regeneration has been investigated for several decades, much work remains to be performed in this field. Future studies should have the following aims: (1) reporting the up-to-date progress in the endogenous cell recruitment strategies; (2) determining the subpopulations of ESPCs, the cellular and molecular mechanisms underlying the migration of these cells and their anti-inflammatory, immunomodulatory and reparative effects; (3) elucidating the chemoattractants that enhance ESPC recruitment and their potential adverse effects; and (4) developing advanced SDDSs for chemoattractant dispatch. Herein, we present a systematic overview of the aforementioned issues to provide a better understanding of endogenous cell recruitment strategies for AC regeneration and repair.


Assuntos
Doenças das Cartilagens , Cartilagem Articular , Células-Tronco Mesenquimais , Humanos , Regeneração , Engenharia Tecidual , Alicerces Teciduais
7.
J Orthop Translat ; 22: 58-66, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32440500

RESUMO

OBJECTIVE: Variation of the solute diffusion within articular cartilage is an important feature of osteoarthritis (OA) progression. For in vitro study of monitoring of the diffusion process, it is essential to simulate physiological conditions as much as possible. Our objective was to investigate the effects of loading patterns on diffusion processes of neutral solutes within osteoarthritic cartilage. METHODS: Osteochondral plugs were harvested from human tibial plateaus and separated into three OA stages according to modified Mankin scoring system. The samples were subjected to static or cyclic compression using a carefully designed loading device. Contrast-enhanced micro-computed tomography (CEµCT) was applied to acquire image sequences while the cartilage was being compressed. The apparent diffusion maps and diffusion coefficients were analysed, as well as histological and stereological assessments of the plugs. RESULTS: The diffusion of neutral solutes was significantly affected by the loading patterns. For OA cartilage with early and middle stages, cyclic loading accelerated contrast agent infiltration compared with static loading. However, for late-stage OA samples, no acceleration of diffusion was observed in the first 2 â€‹h because of the insufficient resilience of compressed cartilage. The accumulation of neutral solutes in an upward invasive fissure also suggested that solutes could penetrate into the fissure under cyclic loading. CONCLUSIONS: To our knowledge, this is the first study to combine the cyclic compression and CEµCT scanning in the diffusion testing of human OA cartilage. This loading pattern could simulate the physiological conditions and reduce the time to reach solute equilibrium within cartilage. The diffusion data may contribute to joint drug-injection therapies for early OA. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: The combination of cyclic loading and CEµCT scanning enabled diffusion analysis of osteoarthritic cartilage under different compressions. A comprehensive evaluation of OA cartilage and subchondral bone may benefit from this technique. The diffusion data provide theoretical support and reference for intra-articular injection of drugs.

8.
J Orthop Translat ; 22: 109-115, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32440506

RESUMO

PURPOSE: The aim of this study was to investigate the location characteristics of cystic lesions in a three-dimensional context and discuss the mechanism of formation. METHODS: A total of 155 femoral head computed tomography images from 94 patients diagnosed with stage II and III osteonecrosis of the femoral head were retrospectively reviewed. Three-dimensional structures of the femoral head including the cystic lesions and necrotic area were reconstructed. We divided each femoral head into eight regions to observe the positional relationship of the cystic lesions, normal areas, and necrotic areas. RESULTS: The regional distribution revealed 14 (13%), 35 (32%), 9 (8%), 25 (23%), 6 (6%), 15 (14%), 4 (4%), and 0 (0%) cystic lesions in regions Ⅰ, Ⅱ, Ⅲ, Ⅳ, Ⅴ, Ⅵ, Ⅶ, and Ⅷ, respectively. The anteromedial zone, A (Ⅰ â€‹+ â€‹â…¢), contained 22% of the lesions, anterolateral zone, B (Ⅱ â€‹+ â€‹â…£), contained 54%, posteromedial zone, C (Ⅴ +Ⅶ), contained 9% of the lesions, and posterolateral zone, D (Ⅵ â€‹+ â€‹â…§), contained 15% of the lesions. Most of the cystic lesions (78%) were located between the normal and necrotic areas; 18% of cystic lesions were in the necrotic area â€‹and 4% were in the normal area. CONCLUSIONS: Cystic lesions most often occur at the junction of the necrotic â€‹and normal areas and are most commonly located in the anterolateral femoral head, which is similar to the distribution of the stress concentration region. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: The study showed the location characteristics of cystic lesions in osteonecrosis of femoral head, which suggested that the formation of cystic lesions may be related to stress and could accelerate the collapse of femoral head. The results can support further research on cystic lesions and provide a reference for doctors' treatment strategies for patients with osteonecrosis of femoral head.

9.
Stem Cell Res Ther ; 11(1): 180, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32430067

RESUMO

BACKGROUND: The dedifferentiation of chondrocytes and the unstable chondrogenic differentiation status of pluripotent mesenchymal stem cells (MSCs) are immense issues in cell-based articular cartilage repair and regenerative strategies. Here, to improve the cartilage characteristics of seed cells, a double biomimetic acellular cartilage extracellular matrix (ACECM)-oriented scaffold was used to mimic the cartilage microenvironment for human umbilical cord Wharton's jelly-derived MSCs (hWJMSCs) and primary cartilage cells (pACs) to regenerate hyaline cartilage. METHODS: A double biomimetic ACECM-oriented scaffold was created from the cartilage extracellular matrix of pig articular cartilage using pulverization decellularization freeze-drying procedures. hWJMSCs and pACs were co-cultured at ratios of 50:50 (co-culture group, ACCC), 0:100 (ACAC group) and 100:0 (ACWJ group) in the ACECM-oriented scaffold, and the co-culture system was implanted in a caprine model for 6 months or 9 months to repair full-thickness articular cartilage defects. The control groups, which had no cells, comprised the blank control (BC) group and the ACECM-oriented scaffold (AC) group. Gross morphology and magnetic resonance imaging (MRI) as well as histological and biomechanical evaluations were used to characterize the cartilage of the repair area. RESULTS: Relative to the control groups, both the gross morphology and histological staining results demonstrated that the neotissue of the ACCC group was more similar to native cartilage and better integrated with the surrounding tissue. Measurements of glycosaminoglycan content and Young's modulus showed that the repair areas had more abundant cartilage-specific content and significantly higher mechanical strength in the ACCC group than in the control groups, especially at 9 months. On MRI, the T2-weighted signal of the repair area was homogeneous, and the oedema signal disappeared almost completely in the ACCC group at 9 months. HLA-ABC immunofluorescence staining demonstrated that hWJMSCs participated in the repair and regeneration of articular cartilage and escaped surveillance and clearance by the caprine immune system. CONCLUSION: The structure and components of double biomimetic ACECM-oriented scaffolds provided a cartilage-like microenvironment for co-cultured seed cells and enhanced the biomechanics and compositions of neotissue. This co-culture system has the potential to overcome the dedifferentiation of passage chondrocytes and the unstable chondrogenic differentiation status of MSCs.


Assuntos
Cartilagem Articular , Animais , Biomimética , Cartilagem Articular/diagnóstico por imagem , Células Cultivadas , Condrócitos , Técnicas de Cocultura , Matriz Extracelular , Cabras , Suínos , Engenharia Tecidual , Alicerces Teciduais
10.
Mol Ther Nucleic Acids ; 18: 841-850, 2019 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-31739209

RESUMO

Osteonecrosis of the femoral head, an intractable but common disease that eventually triggers collapse of the femoral head, is characterized by increased osteoclast activity and markedly decreased osteoblast activity in the necrotic region of the femoral head. MicroRNA (miRNA)-214 (miR-214) may play important roles in vertebrate skeletal development by inhibiting osteoblast function by targeting activating transcription factor 4 (ATF4) and promoting osteoclast function via phosphatase and tensin homolog (PTEN). This study revealed significantly increased levels of miR-214 in necrotic regions, with commensurate changes in the numbers of its target cells (both osteoblasts and osteoclasts). To investigate whether targeting miR-214 could prevent femoral head collapse, we constructed an adeno-associated virus (AAV)-associated anti-miR-214 (AAV-anti-miR-214) and evaluated its function in vivo. AAV-anti-miR-214 promoted osteoblast activity and diminished osteoclast activity, effectively preventing collapse of the femoral head in a rat model of osteonecrosis.

11.
Nanoscale ; 11(42): 19943-19958, 2019 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-31602446

RESUMO

The crosstalk between vascularization and nerve regeneration in the peripheral nervous system has recently been suggested to play an important role in the treatment of peripheral nerve injury. Regenerative strategies via synergistic delivery of multiple biochemical cues have received growing attention, especially the combination of pro-angiogenic factors and neurotrophic factors. Here we developed a self-assembling peptide nanofiber hydrogel dual-functionalized with vascular endothelial growth factor (VEGF)- and brain-derived neurotrophic factor (BDNF)-mimetic peptide epitopes for peripheral nerve reconstruction. It could simultaneously present VEGF- and BDNF-mimetic peptide epitopes and provides a three-dimensional (3D) neurovascular microenvironment for endothelial cell and neural cell growth. In vitro cellular experiments showed that the functionalized peptide hydrogel scaffold effectively promoted the pro-myelination of Schwann cell, as well as the adhesion and proliferation of endothelial cell compared with scaffolds presenting VEGF- or BDNF-mimetic peptide epitope alone. When implanted in a rat model to bridge a critical-size sciatic nerve gap in vivo, the functionalized peptide hydrogel significantly improved the number of newly formed blood vessels, the density of regenerating axons, the morphometric analysis of the regenerated muscles and the electrophysiological findings, indicating the synergistic effect of the two bioactive motifs on peripheral nerve regeneration. Collectively, constructing an artificial neurovascular microenvironment in the lesion area by using the functionalized self-assembling peptide nanofiber hydrogel may have a great potential for promoting nerve tissue engineering and regeneration in other tissues.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Hidrogéis , Regeneração Nervosa/efeitos dos fármacos , Peptídeos , Nervos Periféricos/fisiologia , Fator A de Crescimento do Endotélio Vascular , Animais , Fator Neurotrófico Derivado do Encéfalo/química , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Masculino , Neovascularização Fisiológica/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacologia , Nervos Periféricos/irrigação sanguínea , Ratos , Ratos Sprague-Dawley , Células de Schwann/metabolismo , Alicerces Teciduais/química , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/farmacologia
12.
J Orthop Translat ; 18: 128-141, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31508316

RESUMO

OBJECTIVE: Our study reports the optimization of electrospray human bone marrow stromal cell (hBMSCs)-embedded alginate-gelatin (Alg-Gel, same as following) microspheres for the purpose of their assembly in 3D-printed poly(ε-caprolactone) (PCL) scaffold for the fabrication of a mechanically stable and biological supportive tissue engineering cartilage construct. METHODS: The fabrication of the Alg-Gel microspheres using an electrospray technique was optimized in terms of polydispersity, yield of microspheres and circularity and varying fabrication conditions. PCL scaffolds were designed and printed by melt extrusion. Then, four groups were set: Alg-hBMSC microspheres cultured in the 2D well plate (Alg-hBMSCs+2D) group, Alg-Gel-hBMSC microspheres cultured in the 2D well plate (Alg-Gel-hBMSCs+2D) group, Alg-Gel-hBMSC microspheres embedded in PCL scaffold cultured in the 2D well plate (Alg-Gel-hBMSCs+2D) group and Alg-Gel-hBMSCs microspheres cultured in the 3D bioreactor (Alg-Gel-hBMSCs+3D) group. Cell viability, proliferation and chondrogenic differentiation were evaluated, and mechanical test was performed. RESULTS: Nonaggregated, low polydispersity and almost spherical microspheres of average diameter of 200-300 µm were produced with alginate 1.5 w: v%, gelatin (Type B) concentration of 0.5 w: v % and CaCl2 coagulating bath concentration of 3.0 w: v %, using 30G needle size and 8 kV and 0.6 bar voltage and air pressure, respectively. Alginate with gelatin hydrogel improved viability and promoted hBMSC proliferation better than alginate microspheres. Interestingly, hBMSCs embedded in microspheres assembled in 3D-printed PCL scaffold and cultured in a 3D bioreactor were more proliferative in comparison to the previous two groups (p < 0.05). Similarly, the GAG content, GAG/DNA ratio as well as Coll 2 and Aggr gene expression were increased in the last two groups. CONCLUSION: Optimization of hBMSC-embedded Alg-Gel microspheres produced by electrospray has been performed. The Alg-Gel composition selected allows conservation of hBMSC viability and supports proliferation and matrix deposition. The possibility to seed and assemble microspheres in designed 3D-printed PCL scaffolds for the fabrication of a mechanically stable and biological supportive tissue engineering cartilage construct was demonstrated. TRANSLATIONAL POTENTIAL OF THIS ARTICLE: We optimize and demonstrate that electrospray microsphere fabrication is a cytocompatible and facile process to produce the hBMSC-embedded microsize tissue-like particles that can easily be assembled into a stable construct. This finding could have application in the development of mechanically competent stem cell-based tissue engineering of cartilage regeneration.

13.
Theranostics ; 9(17): 5105-5121, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31410204

RESUMO

Heterogeneity of mesenchymal stem cells (MSCs) influences the cell therapy outcome and the application in tissue engineering. Also, the application of subpopulations of MSCs in cartilage regeneration remains poorly characterized. CD146+ MSCs are identified as the natural ancestors of MSCs and the expression of CD146 are indicative of greater pluripotency and self-renewal potential. Here, we sorted a CD146+ subpopulation from adipose-derived mesenchymal stem cells (ADSCs) for cartilage regeneration. Methods: CD146+ ADSCs were sorted using magnetic activated cell sorting (MACS). Cell surface markers, viability, apoptosis and proliferation were evaluated in vitro. The molecular signatures were analyzed by mRNA and protein expression profiling. By intra-articular injections of cells in a rat osteochondral defect model, we assessed the role of the specific subpopulation in cartilage microenvironment. Finally, CD146+ ADSCs were combined with articular cartilage extracellular matrix (ACECM) scaffold for long term (3, 6 months) cartilage repair. Results: The enriched CD146+ ADSCs showed a high expression of stem cell and pericyte markers, good viability, and immune characteristics to avoid allogeneic rejection. Gene and protein expression profiles revealed that the CD146+ ADSCs had different cellular functions especially in regulation inflammation. In a rat model, CD146+ ADSCs showed a better inflammation-modulating property in the early stage of intra-articular injections. Importantly, CD146+ ADSCs exhibited good biocompatibility with the ACECM scaffold and the CD146+ cell-scaffold composites produced less subcutaneous inflammation. The combination of CD146+ ADSCs with ACECM scaffold can promote better cartilage regeneration in the long term. Conclusion: Our data elucidated the function of the CD146+ ADSC subpopulation, established their role in promoting cartilage repair, and highlighted the significance of cell subpopulations as a novel therapeutic for cartilage regeneration.


Assuntos
Cartilagem Articular/fisiologia , Matriz Extracelular/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Regeneração , Engenharia Tecidual/métodos , Tecido Adiposo/citologia , Animais , Antígeno CD146/genética , Antígeno CD146/metabolismo , Cartilagem Articular/metabolismo , Células Cultivadas , Matriz Extracelular/química , Humanos , Células-Tronco Mesenquimais/metabolismo , Coelhos , Ratos , Ratos Sprague-Dawley , Alicerces Teciduais/química
14.
J Bone Joint Surg Am ; 101(11): 1004-1009, 2019 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-31169577

RESUMO

BACKGROUND: Culture is a key step for detecting periprosthetic joint infection (PJI) before surgery. However, using saline solution lavage and reaspiration in patients with insufficient synovial fluid remains controversial. The objective of this study was to evaluate this technique. METHODS: This study included 286 aspirations performed by 1 surgeon in patients after total joint arthroplasty during the period of April 2015 to August 2018. If >1.0 mL of synovial fluid was obtained, then we directly used the fluid for culture. For cases in which ≤1.0 mL of synovial fluid was aspirated, 10 mL of saline solution was injected and the joint was reaspirated for culture. The samples were injected into 2 blood culture bottles for anaerobic bacterial culture and aerobic bacterial and fungal culture, and were inoculated for 14 days in a BACT/ALERT 3D blood culture system unless microorganisms were detected. A PJI diagnosis was determined on the basis of the modified Musculoskeletal Infection Society criteria. RESULTS: Saline solution lavage and reaspiration were used in 82 cases (47 PJI cases and 35 non-PJI cases), while direct aspiration was used in 204 cases (99 PJI cases and 105 non-PJI cases). The overall rate for the use of saline solution lavage was 28.7% (82 of 286). Among knee cases, the saline solution lavage rate was 15.0% (21 of 140), and among hip cases, the rate was 41.8% (61 of 146). The overall sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) of culture were 0.795 (95% confidence interval [CI], 0.720 to 0.857), 0.957 (95% CI, 0.909 to 0.984), 0.951 (95% CI, 0.896 to 0.982), and 0.817 (95% CI, 0.749 to 0.873); and for "dry tap" cases, they were 0.851 (95% CI, 0.717 to 0.938), 0.857 (95% CI, 0.697 to 0.952), 0.889 (95% CI, 0.760 to 0.963), and 0.811 (95% CI, 0.648 to 0.920), respectively. CONCLUSIONS: Saline solution lavage and reaspiration for culture in patients with insufficient synovial fluid before surgery may be a sound practice. LEVEL OF EVIDENCE: Diagnostic Level III. See Instructions for Authors for a complete description of levels of evidence.


Assuntos
Hemocultura/métodos , Infecções Relacionadas à Prótese/diagnóstico , Líquido Sinovial/metabolismo , Irrigação Terapêutica/métodos , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Infecções Relacionadas à Prótese/metabolismo , Solução Salina/administração & dosagem , Sensibilidade e Especificidade
15.
Medicine (Baltimore) ; 98(18): e15406, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31045795

RESUMO

BACKGROUND: Traumatic vascular injury is caused by explosions and projectiles (bullets and shrapnel); it may affect the arteries and veins of the limbs, and is common in wartime, triggering bleeding, and ischemia. The increasing use of high-energy weapons in modern warfare is associated with severe vascular injuries. METHODS: To summarize the current evidence of diagnosis and treatment for traumatic vascular injury of limbs, for saving limbs and lives, and put forward some new insights, we comprehensively consulted literatures and analyzed progress in injury diagnosis and wound treatment, summarized the advanced treatments now available, especially in wartime, and explored the principal factors in play in an effort to optimize clinical outcomes. RESULTS: Extremity vascular trauma poses several difficult dilemmas in diagnosis and treatment. The increasing use of high-energy weapons in modern warfare is associated with severe vascular injuries. Any delay in treatment may lead to loss of limbs or death. The development of diagnose and treat vascular injury of extremities are the clinical significance to the tip of military medicine, such as the use of fast, cheap, low invasive diagnostic methods, repairing severe vascular injury as soon as possible, using related technologies actively (fasciotomy, etc). CONCLUSION: We point out the frontier of the diagnosis and treatment of traumatic vascular injury, also with a new model of wartime injury treatment in American (forward surgical teams and combat support hospitals), French military surgeons regarding management of war-related vascular wounds and Chinese military ("3 districts and 7 grades" model). Many issues remain to be resolved by further experience and investigation.


Assuntos
Medicina de Emergência/métodos , Extremidades , Medicina Militar/métodos , Lesões do Sistema Vascular/diagnóstico , Lesões do Sistema Vascular/terapia , Amputação Cirúrgica/métodos , Índice Tornozelo-Braço , Traumatismos por Explosões/diagnóstico , Traumatismos por Explosões/terapia , Prótese Vascular , Descompressão Cirúrgica/métodos , Fasciotomia/métodos , Fraturas Ósseas/terapia , Humanos , Militares , Estudos Retrospectivos , Transplante de Pele/métodos , Fatores de Tempo , Índices de Gravidade do Trauma , Estados Unidos , Procedimentos Cirúrgicos Vasculares/métodos , Lesões do Sistema Vascular/diagnóstico por imagem
16.
Biomaterials ; 207: 49-60, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30954885

RESUMO

Flexible nerve guide conduits (NGCs) are desired in peripheral nerve reconstruction near the joints. Inspired by the engineered structure of the helical tube, we addressed the problems of conventional NGCs often mentioned in clinical feedback. We developed a type of helix-flexible NGC (HF-NGC) and evaluated its mechanical properties as well as its flexibility, and it performed excellently during in vitro tests. During the in vivo tests, HF-NGCs and conventional nonflexible NGCs (NF-NGCs) were implanted in a rat sciatic nerve defect model. A short-term investigation showed that Schwan cells (SCs) infiltrated into the helical groove, and most of them belonged to the activated SC type. Compared with NF-NGCs, HF-NGCs had fewer apoptotic SCs. In the long term investigation, HF-NGCs maintained flexibility in vivo after 3 months. Analyses of the morphometric parameters of nerve fibers and the static sciatic index showed that the HF-NGCs had similar regeneration outcomes to those of traditional NF-NGCs. Therefore, this style of HF-NGC could be used to repair peripheral nerve damage in a cross-joint region with less tension during operation and easy to postoperative rehabilitation. We believe that the HF-NGC is a potentially valuable candidate for clinical use.


Assuntos
Regeneração Nervosa/fisiologia , Nervo Isquiático/citologia , Animais , Feminino , Regeneração Tecidual Guiada/métodos , Traumatismos dos Nervos Periféricos/terapia , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/fisiologia , Alicerces Teciduais/química
17.
Stem Cells Int ; 2019: 5130152, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30881462

RESUMO

Seed cells of articular cartilage tissue engineering face many obstacles in their application because of the dedifferentiation of chondrocytes or unstable chondrogenic differentiation status of pluripotent stem cells. To overcome mentioned dilemmas, a simulation of the articular cartilage microenvironment was constructed by primary articular cartilage cells (pACs) and acellular cartilage extracellular matrix- (ACECM-) oriented scaffold cocultured with human umbilical cord Wharton's jelly-derived mesenchymal stem cells (hWJMSCs) in vitro. The coculture groups showed more affluent cartilage special matrix ingredients including collagen II and aggrecan based on the results of histological staining and western blotting and cut down as many pACs as possible. The RT-PCR and cell viability experiments also demonstrated that hWJMSCs were successfully induced to differentiate into chondrocytes when cultured in the simulated cartilage microenvironment, as confirmed by the significant upregulation of collagen II and aggrecan, while the cell proliferation activity of pACs was significantly improved by cell-cell interactions. Therefore, compared with monoculture and chondrogenic induction of inducers, coculture providing a simulated native articular microenvironment was a potential and temperate way to regulate the biological behaviors of pACs and hWJMSCs to regenerate the hyaline articular cartilage.

18.
J Orthop Translat ; 16: 62-70, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30723682

RESUMO

BACKGROUND: The repair of large bone defects remains challenging for orthopaedic surgeons. Bone grafting remains the method of choice; such grafts fill spaces and enhance bone repair. Therapeutic agents also aid bone healing. The objective of this study is to develop a composite bioactive scaffold composed of polylactide-coglycolide (PLGA) and tricalcium phosphate (TCP) (the basic carrier) incorporating osteogenic, bioactive magnesium metal powder (Mg). METHOD: Porous PLGA/TCP scaffolds incorporating Mg were fabricated using a low-temperature rapid-prototyping process. We term the PLGA/TCP/Mg porous scaffold (hereafter, PPS). PLGA/TCP lacking Mg served as the control material when evaluating the efficacy of PPS. A total of 36 New Zealand white rabbits were randomly divided into blank, PLGA/TCP (P/T) and PPS group, with 12 rabbits in each group. We established bone defects 15 mm in length in rabbit radii to evaluate the in vivo osteogenic potential of the bioactive scaffold in terms of the direct controlled release of osteogenic Mg ion during in vivo scaffold degradation. Radiographs of the operated radii were taken immediately after implantation and then at 2, 4, 8 and 12 weeks. Micro-computed tomography of new bone formation and remaining scaffold and histological analysis were performed at 4, 8, 12 weeks after operation. RESULTS: X-ray imaging performed at weeks 4, 8 and 12 post-surgery revealed more newly formed bone within defects implanted with PPS and PLGA/TCP scaffolds than blank group (p < 0.05). And micro-computed tomography performed at weeks 4 and 8 after surgery revealed more newly formed bone within defects implanted with PPS scaffolds than PLGA/TCP scaffolds (p < 0.05). Histologically, the PPS group had more newly mineralized bone than controls (p < 0.05). The increases in new bone areas (total implant regions) in the PPS and PLGA/TCP groups were 19.42% and 5.67% at week 4 and 48.23% and 28.93% at week 8, respectively. The percentages of remaining scaffold material in total implant regions in the PPS and PLGA/TCP groups were 53.30% and 7.65% at week 8 and 20.52% and 2.70% at week 12, respectively. CONCLUSION: Our new PPS composite scaffold may be an excellent orthopaedic substitute; it exhibits good biocompatibility and may potentially have clinical utility. TRANSLATIONAL POTENTIAL OF THIS ARTICLE: Magnesium and beta-tricalcium phosphate had osteoinduction. It is significant to print a novel bone composite scaffold with osteoinduction to repair segmental bone defects. This study evaluated efficacy of PPS in the rabbit radius segmental bone defect model. The results showed that the novel scaffold with good biocompatibility may be an excellent graft and potentially have clinical utility.

19.
Neural Regen Res ; 14(5): 903-912, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30688277

RESUMO

Nerve conduits enhance nerve regeneration in the repair of long-distance peripheral nerve defects. To help optimize the effectiveness of nerve conduits for nerve repair, we developed a multi-step electrospinning process for constructing nerve guide conduits with aligned nanofibers. The alignment of the nerve guide conduits was characterized by scanning electron microscopy and fast Fourier transform. The mechanical performance of the nerve guide conduits was assessed by testing for tensile strength and compression resistance. The biological performance of the aligned fibers was examined using Schwann cells, PC12 cells and dorsal root ganglia in vitro. Immunohistochemistry was performed for the Schwann cell marker S100 and for the neurofilament protein NF200 in PC12 cells and dorsal root ganglia. In the in vivo experiment, a 1.5-cm defect model of the right sciatic nerve in adult female Sprague-Dawley rats was produced and bridged with an aligned nerve guide conduit. Hematoxylin-eosin staining and immunohistochemistry were used to observe the expression of ATF3 and cleaved caspase-3 in the regenerating matrix. The recovery of motor function was evaluated using the static sciatic nerve index. The number of myelinated fibers, axon diameter, fiber diameter, and myelin thickness in the distal nerve were observed by electron microscopy. Gastrocnemius muscle mass ratio was also determined. The analyses revealed that aligned nanofiber nerve guide conduits have good mechanical properties and can induce Schwann cells, PC12 cells and dorsal root ganglia to aggregate along the length of the nanofibers, and promote the growth of longer axons in the latter two (neuronal) cell types. The aligned fiber nerve conduits increased the expression of ATF3 and cleaved caspase-3 at the middle of the regenerative matrix and at the distal nerve segment, improved sciatic nerve function, increased muscle mass of the gastrocnemius muscle, and enhanced recovery of distal nerve ultrastructure. Collectively, the results show that highly aligned nanofibers improve the performance of the nerve conduit bridge, and enhance its effectiveness in repairing peripheral nerve defects.

20.
Theranostics ; 8(18): 5039-5058, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30429885

RESUMO

Even small cartilage defects could finally degenerate to osteoarthritis if left untreated, owing to the poor self-healing ability of articular cartilage. Stem cell transplantation has been well implemented as a common approach in cartilage tissue engineering but has technical complexity and safety concerns. The stem cell homing-based technique emerged as an alternative promising therapy for cartilage repair to overcome traditional limitations. In this study, we constructed a composite hydrogel scaffold by combining an oriented acellular cartilage matrix (ACM) with a bone marrow homing peptide (BMHP)-functionalized self-assembling peptide (SAP). We hypothesized that increased recruitment of endogenous stem cells by the composite scaffold could enhance cartilage regeneration. Methods: To test our hypothesis, in vitro proliferation, attachment and chondrogenic differentiation of rabbit mesenchymal stem cells (MSCs) were tested to confirm the bioactivities of the functionalized peptide hydrogel. The composite scaffold was then implanted into full-thickness cartilage defects on rabbit knee joints for cartilage repair, in comparison with microfracture or other sample groups. Stem cell recruitment was monitored by dual labeling with CD29 and CD90 under confocal microcopy at 1 week after implantation, followed by chondrogenic differentiation examined by qRT-PCR. Repaired tissue of the cartilage defects was evaluated by histological and immunohistochemistry staining, microcomputed tomography (micro-CT) and magnetic resonance imaging (MRI) at 3 and 6 months post-surgery. Macroscopic and histological scoring was done to evaluate the optimal in vivo repair outcomes of this composite scaffold. Results: The functionalized SAP hydrogels could stimulate rabbit MSC proliferation, attachment and chondrogenic differentiation during in vitro culture. At 7 days after implantation, increased recruitment of MSCs based on CD29+ /CD90+ double-positive cells was found in vivo in the composite hydrogel scaffold, as well as upregulation of cartilage-associated genes (aggrecan, Sox9 and type II collagen). After 3 and 6 months post-surgery, the articular cartilage defect in the composite scaffold-treated group was fully covered with cartilage-like tissue with a smooth surface, which was similar to the surrounding native cartilage, according to the results of histological and immunohistochemistry staining, micro-CT and MRI analysis. Macroscopic and histological scoring confirmed that the quality of cartilage repair was significantly improved with implantation of the composite scaffold at each timepoint, in comparison with microfracture or other sample groups. Conclusion: Our findings demonstrated that the composite scaffold could enhance endogenous stem cell homing and chondrogenic differentiation and significantly improve the therapeutic outcome of chondral defects. The present study provides a promising approach for in vivo cartilage repair without cell transplantation. Optimization of this strategy may offer great potential and benefits for clinical application in the future.


Assuntos
Doenças das Cartilagens/terapia , Cartilagem/fisiologia , Condrócitos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Oligopeptídeos/administração & dosagem , Regeneração , Engenharia Tecidual/métodos , Animais , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Modelos Animais de Doenças , Histocitoquímica , Hidrogel de Polietilenoglicol-Dimetacrilato/administração & dosagem , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Coelhos , Resultado do Tratamento , Microtomografia por Raio-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA