Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
J Orthop Translat ; 46: 18-32, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38774916

RESUMO

Background: Osteochondral regeneration has long been recognized as a complex and challenging project in the field of tissue engineering. In particular, reconstructing the osteochondral interface is crucial for determining the effectiveness of the repair. Although several artificial layered or gradient scaffolds have been developed recently to simulate the natural interface, the functions of this unique structure have still not been fully replicated. In this paper, we utilized laser micro-patterning technology (LMPT) to modify the natural osteochondral "plugs" for use as grafts and aimed to directly apply the functional interface unit to repair osteochondral defects in a goat model. Methods: For in vitro evaluations, the optimal combination of LMPT parameters was confirmed through mechanical testing, finite element analysis, and comparing decellularization efficiency. The structural and biological properties of the laser micro-patterned osteochondral implants (LMP-OI) were verified by measuring the permeability of the interface and assessing the recellularization processes. In the goat model for osteochondral regeneration, a conical frustum-shaped defect was specifically created in the weight-bearing area of femoral condyles using a customized trephine with a variable diameter. This unreported defect shape enabled the implant to properly self-fix as expected. Results: The micro-patterning with the suitable pore density and morphology increased the permeability of the LMP-OIs, accelerated decellularization, maintained mechanical stability, and provided two relative independent microenvironments for subsequent recellularization. The LMP-OIs with goat's autologous bone marrow stromal cells in the cartilage layer have securely integrated into the osteochondral defects. At 6 and 12 months after implantation, both imaging and histological assessments showed a significant improvement in the healing of the cartilage and subchondral bone. Conclusion: With the natural interface unit and zonal recellularization, the LMP-OI is an ideal scaffold to repair osteochondral defects especially in large animals. The translational potential of this article: These findings suggest that such a modified xenogeneic osteochondral implant could potentially be explored in clinical translation for treatment of osteochondral injuries. Furthermore, trimming a conical frustum shape to the defect region, especially for large-sized defects, may be an effective way to achieve self-fixing for the implant.

2.
Bioact Mater ; 39: 1-13, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38783924

RESUMO

Irregular articular cartilage injury is a common type of joint trauma, often resulting from intense impacts and other factors that lead to irregularly shaped wounds, the limited regenerative capacity of cartilage and the mismatched shape of the scaffods have contributed to unsatisfactory therapeutic outcomes. While injectable materials are a traditional solution to adapt to irregular cartilage defects, they have limitations, and injectable materials often lack the porous microstructures favorable for the rapid proliferation of cartilage cells. In this study, an injectable porous polyurethane scaffold named PU-BDO-Gelatin-Foam (PUBGF) was prepared. After injection into cartilage defects, PUBGF forms in situ at the site of the defect and exhibits a dynamic microstructure during the initial two weeks. This dynamic microstructure endows the scaffold with the ability to retain substances within its interior, thereby enhancing its capacity to promote chondrogenesis. Furthermore, the chondral repair efficacy of PUBGF was validated by directly injecting it into rat articular cartilage injury sites. The injectable PUBGF scaffold demonstrates a superior potential for promoting the repair of cartilage defects when compared to traditional porous polyurethane scaffolds. The substance retention ability of this injectable porous scaffold makes it a promising option for clinical applications.

3.
Mol Biol Rep ; 50(9): 7437-7444, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37479877

RESUMO

BACKGROUND: We compared the bone microstructure and metabolism of the femoral heads in patients with osteoporosis (OP) and non-OP patients to investigate the pathologic mechanism of OP and guide clinical treatment. METHODS AND RESULTS: From January 2020 to June 2021, we obtained femoral head samples from 30 patients undergoing hip replacement due to femoral neck fracture. All patients were women aged approximately 67 to 80 years (mean age, 74 years). According to the dual-energy X-ray results, the femoral head samples were divided into the OP (T< - 2.5) and non-OP (T > - 1.5) groups. Microcomputed tomography scanning, bone metrology analysis, hematoxylin and eosin staining, and Masson's trichrome staining were used to compare the local bone trabecular microstructure changes. Quantitative reverse transcription PCR was performed to identify changes in the osteogenesis-related genes and the osteoclast-related genes in specific regions to reflect osteogenic and osteoclastic activities. Femoral heads with OP showed significant changes in the local bone microstructure. Bone density, bone volume fraction, and the number and thickness of the bone trabeculae decreased. Local bone metabolism was imbalanced in the areas with microstructural changes in femoral heads with OP, with increased osteoclast activity and decreased osteoblast activity. CONCLUSIONS: Deterioration of bone microstructure is closely related to abnormal bone metabolism associated with the activity of osteoblasts and osteoclasts in osteoporotic femoral heads. Promoting bone formation by improving local bone metabolism, enhancing osteogenic activity and inhibiting osteoclast activity may be a promising way of preventing local OP and osteoporotic fractures.


Assuntos
Cabeça do Fêmur , Osteoporose , Humanos , Feminino , Idoso , Masculino , Cabeça do Fêmur/diagnóstico por imagem , Microtomografia por Raio-X , Osteoclastos , Osteogênese
4.
Interv Neuroradiol ; : 15910199231182850, 2023 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-37309134

RESUMO

BACKGROUND: Clot analogs are essential in animal and in vitro experiments on mechanical thrombectomy devices for treating acute ischemic stroke. Clot analogs should be capable of reproducing a variety of arterial clots observed in clinical practice in terms of histological composition and mechanical properties. METHODS: Bovine blood with added thrombin was stirred in a beaker so that clots could be formed under the condition of dynamic vortical flow. Static clots were also prepared without stirring, and the properties of the static clots and dynamic clots were compared. Histological and scanning electron microscopy experiments were performed. Compression and relaxation tests were performed to evaluate the mechanical properties of the two types of clots. Thromboembolism and thrombectomy tests were conducted in an in vitro circulation model. RESULTS: Compared to the static clots, the dynamic clots prepared under vortical flow displayed a higher fibrin content, and their fibrin network was denser and sturdier than that of the static clots. The stiffness of the dynamic clots was significantly higher than that of the static clots. The stress of both types of clots could decay quickly under large sustained strain. The static clots could break at the bifurcation in the vascular model, while the dynamic clots could be firmly stuck in the vascular model. CONCLUSIONS: Dynamic clots generated in dynamic vortical flow differ significantly from static clots in terms of their composition and mechanical properties, which may be beneficial information for preclinical research on mechanical thrombectomy devices.

5.
Front Bioeng Biotechnol ; 11: 1164252, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37251576

RESUMO

Objective: We investigated the expression of miRNA-214 in human osteoporotic bone tissue and tested the utility of adeno-associated virus (AAV) expressing a miRNA-214 inhibitor in terms of preventing local osteoporosis of the femoral condyle in a rat model of osteoporosis. Methods: (1) Femoral heads of patients who underwent hip replacements at our hospital because of femoral neck fractures were collected and divided into osteoporosis and non-osteoporosis groups based on preoperative bone mineral density data. MiRNA-214 expression was detected in bone tissues exhibiting obvious bone microstructural changes in the two groups. (2) A total of 144 SD female rats were divided into four groups: the Control, Model, Negative control (Model + AAV), and Experimental (Model + anti-miRNA-214) groups. AAV-anti-miRNA-214 was injected locally into the rat femoral condyles; we explored whether this prevented or treated local osteoporosis. Results: (1) MiRNA-214 expression in the human femoral head was significantly increased in the osteoporosis group. (2) Compared to the Model and Model + AAV groups, the bone mineral density (BMD) and femoral condyle bone volume/tissue volume (BV/TV) ratio in the Model + anti-miRNA-214 group were significantly higher; in addition, the number (TB.N) and thickness (TB.Th) of the trabecular bones were increased (all p < 0.05). MiRNA-214 expression in the femoral condyles of the Model + anti-miRNA-214 group was significantly higher than that in the other groups. The expression levels of the osteogenesis-related genes Alp, Bglap, and Col1α1 increased, while those of the osteoclast-related genes NFATc1, Acp5, Ctsk, Mmp9, and Clcn7 decreased. Conclusion: AAV-anti-miRNA-214 promoted osteoblast activity and inhibited osteoclast activity in the femoral condyles of osteoporotic rats, improving bone metabolism and slowing osteoporosis progression.

6.
Commun Biol ; 6(1): 407, 2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-37055517

RESUMO

Mechanical force loading is essential for maintaining bone homeostasis, and unloading exposure can lead to bone loss. Osteoclasts are the only bone resorbing cells and play a crucial role in bone remodeling. The molecular mechanisms underlying mechanical stimulation-induced changes in osteoclast function remain to be fully elucidated. Our previous research found Ca2+-activated Cl- channel Anoctamin 1 (Ano1) was an essential regulator for osteoclast function. Here, we report that Ano1 mediates osteoclast responses to mechanical stimulation. In vitro, osteoclast activities are obviously affected by mechanical stress, which is accompanied by the changes of Ano1 levels, intracellular Cl- concentration and Ca2+ downstream signaling. Ano1 knockout or calcium binding mutants blunts the response of osteoclast to mechanical stimulation. In vivo, Ano1 knockout in osteoclast blunts loading induced osteoclast inhibition and unloading induced bone loss and. These results demonstrate that Ano1 plays an important role in mechanical stimulation induced osteoclast activity changes.


Assuntos
Canais de Cloreto , Osteoclastos , Anoctamina-1/genética , Anoctamina-1/metabolismo , Canais de Cloreto/genética , Osteoclastos/metabolismo , Transdução de Sinais/fisiologia
7.
Int J Biol Macromol ; 235: 123861, 2023 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-36870644

RESUMO

Compared with most nondegradable or slowly degradable bone repair materials, bioactive biodegradable porous scaffolds with certain mechanical strengths can promote the regeneration of both new bone and vasculature while the cavity created by their degradation can be replaced by the infiltration of new bone tissue. Mineralized collagen (MC) is the basic structural unit of bone tissue, and silk fibroin (SF) is a natural polymer with adjustable degradation rates and superior mechanical properties. In this study, a three-dimensional porous biomimetic composite scaffold with a two-component SF-MC system was constructed based on the advantages of both materials. The spherical mineral agglomerates of the MC were uniformly distributed on the surface and inside the SF skeleton, which ensured good mechanical properties while regulating the degradation rate of the scaffold. Second, the SF-MC scaffold had good osteogenic induction of bone marrow mesenchymal stem cells (BMSCs) and preosteoblasts (MC3T3-E1) and also promoted the proliferation of MC3T3-E1 cells. Finally, in vivo 5 mm cranial defect repair experiments confirmed that the SF-MC scaffold stimulated vascular regeneration and promoted new bone regeneration in vivo by means of in situ regeneration. Overall, we believe that this low-cost biomimetic biodegradable SF-MC scaffold with many advantages has some clinical translation prospects.


Assuntos
Fibroínas , Ratos , Animais , Fibroínas/farmacologia , Fibroínas/química , Alicerces Teciduais/química , Engenharia Tecidual/métodos , Regeneração Óssea/fisiologia , Osteogênese , Colágeno , Crânio , Seda/química
8.
Bioact Mater ; 26: 370-386, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36942011

RESUMO

Autologous nerve grafting serves is considered the gold standard treatment for peripheral nerve defects; however, limited availability and donor area destruction restrict its widespread clinical application. Although the performance of allogeneic decellularized nerve implants has been explored, challenges such as insufficient human donors have been a major drawback to its clinical use. Tissue-engineered neural regeneration materials have been developed over the years, and researchers have explored strategies to mimic the peripheral neural microenvironment during the design of nerve catheter grafts, namely the extracellular matrix (ECM), which includes mechanical, physical, and biochemical signals that support nerve regeneration. In this study, polycaprolactone/silk fibroin (PCL/SF)-aligned electrospun material was modified with ECM derived from human umbilical cord mesenchymal stem cells (hUMSCs), and a dual-bionic nerve regeneration material was successfully fabricated. The results indicated that the developed biomimetic material had excellent biological properties, providing sufficient anchorage for Schwann cells and subsequent axon regeneration and angiogenesis processes. Moreover, the dual-bionic material exerted a similar effect to that of autologous nerve transplantation in bridging peripheral nerve defects in rats. In conclusion, this study provides a new concept for designing neural regeneration materials, and the prepared dual-bionic repair materials have excellent auxiliary regenerative ability and further preclinical testing is warranted to evaluate its clinical application potential.

9.
ACS Nano ; 17(7): 6373-6386, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36961738

RESUMO

Magnetic biomaterials are widely used in the field of tissue engineering because of their functions such as drug delivery and targeted therapy. In this study, a magnetically responsive composite microcarrier was prepared through in situ polymerization of dopamine with Fe3O4 (MS) to form a complex. The magnetic composite microcarriers are paramagnetic and have certain magnetic responsiveness, suitable pore size porosity for cell growth, and good blood compatibility and biocompatibility. The bone marrow mesenchyml stem cells (BMSCs) were cultured on magnetic composite microcarriers, and a static magnetic field (SMF) was applied. The results showed that BMSCs adhered to the microcarriers proliferated under the action of horizontal and vertical forces. Magnetic composite microcarriers loaded with BMSCs were implanted into the SD rat model of cartilage defect, and a magnet was added to the operative side. After 12 weeks, cartilage regeneration was observed. The results of gross observation and histological immunostaining 1 month, 2 months, and 3 mounths after operation showed that the magnetic composite microcarriers of loaded cells promoted the early maturation of cartilage and collagen secretion, and the effect of cartilage repair was significantly better than that of the control group. Gait analysis showed that implanting magnetic composite microcarriers loaded with stem cells can reduce postoperative pain and promote limb recovery in SD rats. In conclusion, this study suggests that magnetic composite microcarriers are promising tissue-engineered scaffolds for cartilage regeneration and repair.


Assuntos
Cartilagem Articular , Células-Tronco Mesenquimais , Ratos , Animais , Ratos Sprague-Dawley , Alicerces Teciduais , Engenharia Tecidual/métodos , Proliferação de Células , Fenômenos Magnéticos , Células Cultivadas
10.
Artigo em Inglês | MEDLINE | ID: mdl-36758166

RESUMO

Inflammation is part of the natural healing response, but persistent inflammatory events tend to contribute to pathology changes of tendon or ligament. Phenotypic switching of macrophages within the inflammatory niche is crucial for tendon healing. One viable strategy to improve the functional and biomechanical properties of ruptured tendons is to modulate the transition from inflammatory to regenerative signals during tendon regeneration at the site of injury. Here, we developed a tendon repair scaffold made of biodegradable polycaprolactone by electrospinning, which was modified to deliver Wnt3a protein and served as an implant to improve tendon healing in a rat model of Achilles tendon defect. During the in vitro study, Wnt3a protein promoted the polarization of M2 macrophages. In the in vivo experiment, Wnt3a scaffold promoted the early recruitment and counting curve of macrophages and increased the proportion of M2 macrophages. Achilles function index and mechanical properties showed that the implantation effect of the Wnt3a group was better than that of the control group. We believe that this type of scaffold can be used to repair tendon defects. This work highlights the beneficial role of local delivery of biological factors in directing inflammatory responses toward regenerative strategies in tendon healing.

11.
J Orthop Translat ; 37: 126-142, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36313533

RESUMO

Background: Osteonecrosis of the femoral head (ONFH) is a refractory disease due to its unclear pathomechanism. Therapies during the early stage of ONFH have not achieved satisfactory results. Therefore, this study aims to explore the available evidence for the therapeutic effect of human umbilical cord mesenchymal stem cells (HUCMSCs) on early-stage traumatic ONFH. Methods: Early-stage traumatic ONFH was established. The femoral heads of rats were then locally administered HUCMSCs. Four weeks and eight weeks after surgery, bone repair of the necrotic area in the femoral head was analyzed to evaluate the therapeutic effect of HUCMSCs using micro-CT, histopathological staining, immunofluorescence staining, Luminex. Results: HUCMSCs were still present in the femoral head four weeks later, and the morphological, micro-CT and histopathological outcomes in the 4-week HUCMSC-treated group were better than those in the model, NS and 8-week HUCMSC-treated groups. Local transplantation of HUCMSCs promoted bone repair and prevented bone loss in the necrotic area of the femoral head. Conclusions: HUCMSCs can survive and positively affect the femoral head through local transplantation in early-stage traumatic ONFH. The conclusions of this study can provide a treatment option for patients who have ONFH and can serve as basic research on the advanced development of this disease. The Translational potential of this article: The study indicated that the positive effect of exogenous HUCMSCs in the treatment of early-stage traumatic ONFH provides the solid basis and guidance for the clinical application of HUCMSCs.

12.
Orthop Surg ; 14(9): 1951-1957, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35924692

RESUMO

Osteonecrosis of the femoral head (ONFH) is a progressive disease that often necessitates hip replacement if hip preservation therapy fails. ONFH places a heavy economic burden and severe psychological pressure on patients. At present, ONFH is treated by either surgical or non-surgical methods. In clinical practice, stem cells combined with surgery has achieved some positive results, but many problems remain to be resolved. Exosomes are small vesicles of 30-150 nm, which are rich in various nucleic acids, proteins, and small molecules depending on the cells from which they are derived. A growing number of studies have found that exosomes play an important role in tissue damage repair. In comparison with stem cells, exosomes have lower immunogenicity. Also, exosomes can promote cell proliferation and inhibit tumor growth. In addition, exosomes can also be used as natural carriers of drugs. Many studies have shown that exosomes have therapeutic effects in hormone-induced ONFH. Exosomes have the effect of promoting vascular regeneration and show good application prospects in ONFH. Here, we present a review of studies on the application of exosomes in ONFH to provide a reference for future research.


Assuntos
Exossomos , Necrose da Cabeça do Fêmur , Ácidos Nucleicos , Osteonecrose , Exossomos/metabolismo , Cabeça do Fêmur , Necrose da Cabeça do Fêmur/induzido quimicamente , Hormônios/efeitos adversos , Hormônios/metabolismo , Humanos , Ácidos Nucleicos/efeitos adversos , Ácidos Nucleicos/metabolismo
13.
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.

14.
Orthop Surg ; 14(5): 831-839, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35445585

RESUMO

Osteonecrosis of the femoral head (ONFH) is a crippling disease which is due to a lack of effective therapeutic measures. Its natural progression is rapid, the internal bone structure of the femoral head changes dramatically, and the subsequent fractures and collapse cause severe hip pain and loss of hip function. Femoral head collapse is a critical turning point in the development of ONFH and is related to the prognosis of patients. Early prevention and intervention help to preserve the hip joint and delay femoral head collapse. However, the mechanism of collapse still needs to be further studied because it is affected by different complex factors. This review discusses the underlying causes of femoral head collapse from two aspects: structural degradation and regional changes of biomechanical properties in the necrotic femoral head.


Assuntos
Necrose da Cabeça do Fêmur , Cabeça do Fêmur , Necrose da Cabeça do Fêmur/etiologia , Necrose da Cabeça do Fêmur/patologia , Quadril , Articulação do Quadril , Humanos , Imageamento por Ressonância Magnética
15.
Bioact Mater ; 18: 300-320, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35387172

RESUMO

Compared to conventional artificial nerve guide conduits (NGCs) prepared using natural polymers or synthetic polymers, acellular nerve grafts (ACNGs) derived from natural nerves with eliminated immune components have natural bionic advantages in composition and structure that polymer materials do not have. To further optimize the repair effect of ACNGs, in this study, we used a composite technology based on supercritical carbon dioxide (scCO2) extraction to process the peripheral nerve of a large mammal, the Yorkshire pig, and obtained an innovative Acellular nerve xenografts (ANXs, namely, CD + scCO2 NG). After scCO2 extraction, the fat and DNA content in CD + scCO2 NG has been removed to the greatest extent, which can better supported cell adhesion and proliferation, inducing an extremely weak inflammatory response. Interestingly, the protein in the CD + scCO2 NG was primarily involved in signaling pathways related to axon guidance. Moreover, compared with the pure chemical decellularized nerve graft (CD NG), the DRG axons grew naturally on the CD + scCO2 NG membrane and extended long distances. In vivo studies further revealed that the regenerated nerve axons had basically crossed the CD + scCO2 NG 3 weeks after surgery. 12 weeks after surgery, CD + scCO2 NG was similar to autologous nerves in improving the quality of nerve regeneration, target muscle morphology and motor function recovery and was significantly better than hollow NGCs and CD NG. Therefore, we believe that the fully decellularized and fat-free porcine ACNGs may be the most promising "bridge" for repairing human nerve defects at this stage and for some time to come.

16.
Bioact Mater ; 17: 471-487, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35415294

RESUMO

Effective strategy of hemostasis and promoting angiogenesis are becoming increasingly urgent in modern medicine due to millions of deaths caused by tissue damage and inflammation. The tissue adhesive has been favored as an optimistic and efficient path to stop bleeding, while, current adhesive presents limitations on wound care or potential degradation safety in clinical practice. Therefore, it is of great clinical significance to construct multifunctional wound adhesive to address the issues. Based on pro-angiogenic property of l-Arginine (L-Arg), in this study, the novel tissue adhesive (G-DLPUs) constructed by L-Arg-based degradable polyurethane (DLPU) and GelMA were prepared for wound care. After systematic characterization, we found that the G-DLPUs were endowed with excellent capability in shape-adaptive adhesion. Moreover, the L-Arg released and the generation of NO during degradation were verified which would enhance wound healing. Following the in vivo biocompatibility was verified, the hemostatic effect of the damaged organ was tested using a rat liver hemorrhage model, from which reveals that the G-DLPUs can reduce liver bleeding by nearly 75% and no obvious inflammatory cells observed around the tissue. Moreover, the wound care effect was confirmed in a mouse full-thickness skin defect model, showing that the hydrogel adhesive significantly improves the thickness of newly formed dermis and enhance vascularization (CD31 staining). In summary, the G-DLPUs are promising candidate to act as multifunctional wound care adhesive for both damaged organ and trauma.

17.
J Orthop Translat ; 33: 13-23, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35198379

RESUMO

BACKGROUND: /Objective: The treatment of bone defect has always been a difficult problem in orthopedic clinic. The search for alternative biodegradable implants is a hot topic. The development of biodegradable magnesium scaffolds for the treatment of bone defects has long been a goal of the public. METHODS: In this study, we proposed a porous magnesium scaffold prepared by 3D gel printing and surface modification with an additional calcium phosphate coating and use of its strength, degradability and slow degradation rate in a bone graft substitute material. The porous magnesium granular scaffold was prepared by 3D gel printing technology and modified by DCPD (Dibasic Calcium Phosphate Dihydrate) coating. The biocompatibility, degradation rate, and osteogenic ability of the scaffold were evaluated in vitro and in vivo. RESULTS: The biocompatibility, in vivo degradation and bone defect healing response of the implants were investigated. Porous magnesium scaffolds were successfully prepared, and the strength of sintered scaffolds reached 5.38 â€‹MPa. The degradation rates of scaffolds were significantly reduced after coating with DCPD. The cell compatibility evaluation showed that DCPD-coated Mg scaffold was suitable for cell proliferation. In vivo biosafety monitoring showed that scaffold implantation did not cause an increase in Mg ion concentration in vivo, and no toxic damage was detected in the liver or kidney. Micro-CT and pathological results showed that a large amount of new bone was formed at 6 weeks. At 12 weeks, approximately 52% of the scaffold volume remained. At 24 weeks, osteogenesis, which was stimulated by some residual scaffold, still can be observed. In summary, this study suggests that 3D gel-printed DCPD-coated porous magnesium scaffolds have great potential as bone graft alternatives. CONCLUSION: In summary, this study suggests that 3D gel-printed DCPD-coated porous magnesium scaffolds have great potential as bone graft alternatives. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: The translational potential of this article is to make use of the advantages of 3D gel printing technology with higher efficiency and lower cost compared with SLM and SLS technologies, and use pure magnesium powder as raw material to prepare degradable porous magnesium metal scaffolds, opening up a new technical route for the preparation of degradable porous magnesium scaffolds which are made for bone defect regeneration in the future.

18.
Carbohydr Polym ; 281: 119044, 2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35074117

RESUMO

Biocompatible and electroactive biomaterials have good potential on peripheral nerve repair. Bacterial cellulose (BC) shows excellent biocompatibility and is easy to modified, however it lacks electroactivity. In this study, biocompatible, conductive, and transparent bacterial cellulose/poly(3,4-ethylenedioxythiophene)-sulfonated nanofibers (BC/PEDOT-SNFs, BPS) composite membranes were prepared through the in-situ polymerization of PEDOT and the doping of SNFs. The polymerization of PEDOT endowed BC with conductivity, making the BPS membranes conducive to the adhesion and proliferation of adipose-derived stem cells (ADSCs). The conductivity of BPS was affected by the SNFs doped, and its value was up to 1.8 × 10-2 S/cm while the sulfonation degree of SNFs reached 93%. Furthermore, nerve conduits made of BPS were implanted in-vivo for 12 weeks, and it great improved the peripheral nerve repair effect. In summary, BPS membranes with excellent conductivity and multiple merits for cells loading, hold great application potential for peripheral nerve repair.


Assuntos
Nanofibras , Compostos Bicíclicos Heterocíclicos com Pontes , Celulose , Nervos Periféricos , Polímeros
19.
J Orthop Translat ; 30: 112-121, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34722154

RESUMO

BACKGROUND/OBJECTIVE: We seek to figure out the effect of stable and powerful mechanical microenvironment provided by Ti alloy as a part of subchondral bone scaffold on long-term cartilage regeneration.Methods: we developed a bilayered osteochondral scaffold based on the assumption that a stiff subchondral bony compartment would provide stable mechanical support for cartilage regeneration and enhance subchondral bone regeneration. The subchondral bony compartment was prepared from 3D printed Ti alloy, and the cartilage compartment was created from a freeze-dried collagen sponge, which was reinforced by poly-lactic-co-glycolic acid (PLGA). RESULTS: In vitro evaluations confirmed the biocompatibility of the scaffold materials, while in vivo evaluations demonstrated that the mechanical support provided by 3D printed Ti alloy layer plays an important role in the long-term regeneration of cartilage by accelerating osteochondral formation and its integration with the adjacent host tissue in osteochondral defect model at rabbit femoral trochlea after 24 weeks. CONCLUSION: Mechanical support provided by 3D printing Ti alloy promotes cartilage regeneration by promoting subchondral bone regeneration and providing mechanical support platform for cartilage synergistically. TRANSLATIONAL POTENTIAL STATEMENT: The raw materials used in our double-layer osteochondral scaffolds are all FDA approved materials for clinical use. 3D printed titanium alloy scaffolds can promote bone regeneration and provide mechanical support for cartilage regeneration, which is very suitable for clinical scenes of osteochondral defects. In fact, we are conducting clinical trials based on our scaffolds. We believe that in the near future, the scaffold we designed and developed can be formally applied in clinical practice.

20.
Orthop Surg ; 13(6): 1843-1852, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34664417

RESUMO

OBJECTIVE: To investigate the efficacy and safety of core decompression (CD) with local administration of zoledronate and enriched bone marrow mononuclear cells (BMMCS) for the treatment of non-traumatic osteonecrosis of femoral head (ONFH). METHODS: A total of 17 patients (30 hips) diagnosed with stage II and III ONFH according to the 2019 revised Association for Research on Osseous Circulation (ARCO) staging criteria from 2012 to 2014 were retrospectively reviewed. The patients received the following therapy: the BMMCs and zoledronate were injected into the necrotic zone, respectively, along with CD. The mean age of the patients was 36.8 years; 14 were men and three were women. All patients included had non-traumatic ONFH and a minimum follow-up of 5 years, which ended when total hip arthroplasty (THA) was performed. Imaging modalities, including plain radiography, computed tomography (CT), and magnetic resonance imaging (MRI) were taken pre- and postoperatively. Harris hip score (HHS) was used to evaluate the functional outcomes of femoral head necrosis. Kaplan-Meier analysis was adopted to determine the probability of survivorship with THA as the end point in this series of patients. The correlation between radiological progression or THA and related risk factors were further analyzed. All complications were recorded. RESULTS: With THA as the follow-up endpoint, All patients were followed up for an average of 69.1 ± 20.5 months (range, 18-95 months). Preoperative imaging found six hips (20%) at ARCO stage II, 14 hips (46.7%) at stage IIIA, 10 hips (33.3%) at stage IIIB. Fourteen hips (46.7%) shown progression radiologically, while six hips (20%) underwent TKA among these patients with hip preservation. The cumulative survival was 80% (95% CI, 0.608-905) at 5 years with THA as the end point. HHS improved from 63.3 ± 8.7 preoperatively to 74.6 ± 20.6 postoperatively (P = 0.000). Radiological progression was found to be associated with ARCO stage, Japanese Investigation Committee (JIC) type, and corticosteroid exposure (P = 0.047; P = 0.012; P = 0.031). However, no correlation was found between conversion to THA and the known risk factors. No major complication was reported, with only four patients complaining about general weakness and muscle soreness, and all disappeared within 2-3 days. CONCLUSIONS: The novel treatment modality could relieve pain, delay the progression of collapse, which might be an effective and safe method for hip preservation of early and mid-term ONFH. However, the effect of this method may be related to ARCO stage, JIC type, and corticosteroid exposure.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Descompressão Cirúrgica/métodos , Necrose da Cabeça do Fêmur/terapia , Ácido Zoledrônico/administração & dosagem , Adulto , Conservadores da Densidade Óssea/administração & dosagem , Feminino , Humanos , Injeções Intralesionais , Masculino , Pessoa de Meia-Idade , Medidas de Resultados Relatados pelo Paciente , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA