Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38411502

RESUMO

Rotator cuff tear (RCT) is the most common cause of disability in the upper-extremity.1 It results in 4.5 million physician visits in the United States every year and is the most common etiology of shoulder conditions evaluated by orthopedic surgeons.2,3 Over 460,000 RCT repair surgeries are performed in the United States annually.4 Rotator cuff (RC) retear and failure to heal remain significant post-operative complications.5 Literature suggests that the retear rates can range from 29.5% to as high as 94%.6,7 Weakened and irregular enthesis regeneration is a crucial factor in post-surgical failure.8 Although commercially available RC repair grafts have been introduced to augment RC enthesis repair, they have been associated with mixed clinical outcomes.9,10 These grafts lack appropriate biological cues such as stem cells and signaling molecules at the bone-tendon interface. Additionally, they do little to prevent fibrovascular scar tissue formation, which causes the RC to be susceptible to retear. Advances in tissue engineering have demonstrated that mesenchymal stem cells (MSCs) and growth factors (GFs) enhance RC enthesis regeneration in animal models. These models show that delivering MSCs and GFs to the site of RC tear enhances native enthesis repair and leads to greater mechanical strength. Additionally, these models demonstrate that MSCs and GFs may be delivered through a variety of methods including direct injection, saturation of repair materials, and loaded microspheres. Grafts that incorporate MSCs and GFs enhance anti-inflammation, osteogenesis, angiogenesis, and chondrogenesis in the RC repair process. It is crucial that the techniques which have shown success in animal models are incorporated into the clinincal setting. A gap currently exists between the promising biological factors which have been investigated in animal models and the RC repair grafts that can be used in the clinical setting. Future RC repair grafts must allow for stable implantation and fixation, be compatible with current arthroscopic techniques, and have the capability to deliver MSCs and/or GF. References (Full citations include in manuscript) 1.Kovacevic (2020) 2. Moran (2023) 3. Piper (2018) 4. IData (2018) 5. Yamaura (2023) 6. Park (2021) 7. Davey (2023) 8. Smietana (2017) 9. Walton (2007) 10. Soler (2007).

2.
J Biomed Mater Res B Appl Biomater ; 112(1): e35360, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38247252

RESUMO

Nontraumatic osteonecrosis of the femoral head (ONFH) is a refractory condition that commonly results in femoral head collapse and degenerative arthritis of the hip. In the early stages, surgical procedures for hip preservation, including core decompression (CD), have been developed to prevent progressive collapse of the femoral head. Optimization of bone regeneration and biological augmentation may further enhance the therapeutic efficacy of CD for ONFH. Thus, combining CD with cell-based therapy has recently been proposed. In fact, patients treated with cell-based therapy using autologous bone marrow concentrate demonstrate improved survivorship of the femoral head, compared with conventional CD alone. Preclinical research studies to investigate adjunctive therapies for CD often utilize the rabbit model of corticosteroid-induced ONFH. Mesenchymal stem cells (MSCs) are known to promote osteogenesis and angiogenesis, and decrease inflammation in bone. Local drug delivery systems have the potential to achieve targeted therapeutic effects by precisely controlling the drug release rate. Scaffolds can provide an osteoconductive structural framework to facilitate the repair of osteonecrotic bone tissue. We focused on the combination of both cell-based and scaffold-based therapies for bone tissue regeneration in ONFH. We hypothesized that combining CD and osteoconductive scaffolds would provide mechanical strength and structural cell guidance; and that combining CD and genetically modified (GM) MSCs to express relevant cytokines, chemokines, and growth factors would promote bone tissue repair. We developed GM MSCs that overexpress the anti-inflammatory, pro-reconstructive cytokines platelet-derived growth factor-BB to provide MSCs with additional benefits and investigated the efficacy of combinations of these GM MSCs and scaffolds for treatment of ONFH in skeletally mature male New Zealand white rabbits. In the future, the long-term safety, efficacy, durability, and cost-effectiveness of these and other biological and mechanical treatments must be demonstrated for the patients affected by ONFH.


Assuntos
Cabeça do Fêmur , Procedimentos Ortopédicos , Humanos , Animais , Masculino , Coelhos , Corticosteroides , Regeneração Óssea , Citocinas
3.
Nat Commun ; 14(1): 4455, 2023 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-37488113

RESUMO

Bone transport is a surgery-driven procedure for the treatment of large bone defects. However, challenging complications include prolonged consolidation, docking site nonunion and pin tract infection. Here, we develop an osteoinductive and biodegradable intramedullary implant by a hybrid tissue engineering construct technique to enable sustained delivery of bone morphogenetic protein-2 as an adjunctive therapy. In a male rat bone transport model, the eluting bone morphogenetic protein-2 from the implants accelerates bone formation and remodeling, leading to early bony fusion as shown by imaging, mechanical testing, histological analysis, and microarray assays. Moreover, no pin tract infection but tight osseointegration are observed. In contrast, conventional treatments show higher proportion of docking site nonunion and pin tract infection. The findings of this study demonstrate that the novel intramedullary implant holds great promise for advancing bone transport techniques by promoting bone regeneration and reducing complications in the treatment of bone defects.


Assuntos
Implantes Absorvíveis , Osteogênese , Masculino , Animais , Ratos , Bioensaio , Regeneração Óssea , Osseointegração
4.
Int J Bioprint ; 9(4): 705, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37323480

RESUMO

Bioink preparation is an important yet challenging step for bioprinting with hydrogels, as it involves fast and homogeneous mixing of various viscous components. In this study, we have developed an automated active mixing platform (AAMP), which allows for high-quality preparation of hydrogel bioinks. The design of AAMP, adapted from syringe pumps, provides many advantages, including low cost, automated control, high precision, customizability, and great cytocompatibility, as well as the potential to intelligently detect the homogeneity. To demonstrate the capability of AAMP, mixing of different hydrogel components, including alginate and xanthan gum with and without Ca2+, alginate and Laponite, PEGDMA and xanthan gum, was performed to investigate an alginate hydrogel preparation process. Colorimetric analyses were carried out to evaluate the mixing outcome with AAMP. Result showed that AAMP can prepare homogeneous hydrogel mixing in a fast and automated fashion. A multiphysics COMSOL simulation is carried out to further validate the results. Moreover, cell viability and proliferation study were performed in a cell encapsulation mixing experiment to validate the cytocompatibility of the AAMP. The AAMP has demonstrated great capability in hydrogel bioink preparation and could therefore holds great promise and wide applications in bioprinting and tissue engineering.

5.
Stem Cell Res Ther ; 14(1): 99, 2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-37085909

RESUMO

BACKGROUND: Continuous cross talk between MSCs and macrophages is integral to acute and chronic inflammation resulting from contaminated polyethylene particles (cPE); however, the effect of this inflammatory microenvironment on mitochondrial metabolism has not been fully elucidated. We hypothesized that (a) exposure to cPE leads to impaired mitochondrial metabolism and glycolytic reprogramming and (b) macrophages play a key role in this pathway. METHODS: We cultured MSCs with/without uncommitted M0 macrophages, with/without cPE in 3-dimensional gelatin methacrylate (3D GelMA) constructs/scaffolds. We evaluated mitochondrial function (membrane potential and reactive oxygen species-ROS production), metabolic pathways for adenosine triphosphate (ATP) production (glycolysis or oxidative phosphorylation) and response to stress mechanisms. We also studied macrophage polarization toward the pro-inflammatory M1 or the anti-inflammatory M2 phenotype and the osteogenic differentiation of MSCs. RESULTS: Exposure to cPE impaired mitochondrial metabolism of MSCs; addition of M0 macrophages restored healthy mitochondrial function. Macrophages exposed to cPE-induced glycolytic reprogramming, but also initiated a response to this stress to restore mitochondrial biogenesis and homeostatic oxidative phosphorylation. Uncommitted M0 macrophages in coculture with MSC polarized to both M1 and M2 phenotypes. Osteogenesis was comparable among groups after 21 days. CONCLUSION: This work confirmed that cPE exposure triggers impaired mitochondrial metabolism and glycolytic reprogramming in a 3D coculture model of MSCs and macrophages and demonstrated that macrophages cocultured with MSCs undergo metabolic changes to maintain energy production and restore homeostatic metabolism.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Polietileno/metabolismo , Polietileno/farmacologia , Macrófagos/metabolismo , Metaboloma , Células-Tronco Mesenquimais/metabolismo
6.
J Biomed Mater Res A ; 111(8): 1120-1134, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36606330

RESUMO

Core decompression (CD) with mesenchymal stromal cells (MSCs) is an effective therapy for early-stage osteonecrosis of the femoral head (ONFH). Preconditioning of MSCs, using inflammatory mediators, is widely used in immunology and various cell therapies. We developed a three-dimensional printed functionally graded scaffold (FGS), made of ß-TCP and PCL, for cell delivery at a specific location. The present study examined the efficacy of CD treatments with genetically modified (GM) MSCs over-expressing PDGF-BB (PDGF-MSCs) or GM MSCs co-over-expressing IL-4 and PDGF-BB and preconditioned for three days of exposure to lipopolysaccharide and tumor necrosis factor-alpha (IL-4-PDGF-pMSCs) using the FGS for treating steroid-induced ONFH in rabbits. We compared CD without cell-therapy, with IL-4-PDGF-pMSCs alone, and with FGS loaded with PDGF-MSCs or IL-4-PDGF-pMSCs. For the area inside the CD, the bone volume in the CD alone was higher than in both FGS groups. The IL-4-PDGF-pMSCs alone and FGS + PDGF-MSCs reduced the occurrence of empty lacunae and improved osteoclastogenesis. There was no significant difference in angiogenesis among the four groups. The combined effect of GM MSCs or pMSCs and the FGS was not superior to the effect of each alone. To establish an important adjunctive therapy for CD for early ONFH in the future, it is necessary and essential to develop an FGS that delivers biologics appropriately and provides structural and mechanical support.


Assuntos
Células-Tronco Mesenquimais , Osteonecrose , Animais , Coelhos , Cabeça do Fêmur/patologia , Cabeça do Fêmur/cirurgia , Becaplermina , Interleucina-4/farmacologia , Regeneração Óssea , Células-Tronco Mesenquimais/patologia , Corticosteroides/farmacologia , Osteonecrose/induzido quimicamente , Osteonecrose/terapia , Osteonecrose/patologia
7.
Bioact Mater ; 19: 167-178, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35510174

RESUMO

Conventional synthetic vascular grafts are associated with significant failure rates due to their mismatched mechanical properties with the native vessel and poor regenerative potential. Though different tissue engineering approaches have been used to improve the biocompatibility of synthetic vascular grafts, it is still crucial to develop a new generation of synthetic grafts that can match the dynamics of native vessel and direct the host response to achieve robust vascular regeneration. The size of pores within implanted biomaterials has shown significant effects on macrophage polarization, which has been further confirmed as necessary for efficient vascular formation and remodeling. Here, we developed biodegradable, autoclavable synthetic vascular grafts from a new polyurethane elastomer and tailored the grafts' interconnected pore sizes to promote macrophage populations with a pro-regenerative phenotype and improve vascular regeneration and patency rate. The synthetic vascular grafts showed similar mechanical properties to native blood vessels, encouraged macrophage populations with varying M2 to M1 phenotypic expression, and maintained patency and vascular regeneration in a one-month rat carotid interposition model and in a four-month rat aortic interposition model. This innovative bioactive synthetic vascular graft holds promise to treat clinical vascular diseases.

8.
Biomaterials ; 293: 121969, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36566553

RESUMO

Gelatin methacryloyl (GelMA)/alginate-based hydrogels have shown great promise in bioprinting, but their printability is limited at room temperature. In this paper, we present our development of a room temperature printable hydrogel bioink by introducing polyethylene glycol dimethacrylate (PEGDMA) and xanthan gum into the GelMA/alginate system. The inclusion of PEGDMA facilitates tuning of the hydrogel's mechanical property, while xanthan gum improves the viscosity of the hydrogel system and allows easy extrusion at room temperature. To fine-tune the mechanical and degradation properties, methacrylated xanthan gum was synthesized and chemically crosslinked to the system. We systematically characterized this hydrogel with attention to printability, strut size, mechanical property, degradation and cytocompatibility, and achieved a broad range of compression modulus (∼10-100 kPa) and degradation profile (100% degradation by 24 h-40% by 2 weeks). Moreover, xanthan gum demonstrated solubility in ionic solutions such as cell culture medium, which is essential for biocompatibility. Live/dead staining showed that cell viability in the printed hydrogels was over 90% for 7 days. Metabolic activity analysis demonstrated excellent cell proliferation and survival within 4 weeks of incubation. In summary, the newly developed hydrogel system has demonstrated distinct features including extrusion printability, widely tunable mechanical property and degradation, ionic solubility, and cytocompatibility. It offers great flexibility in bioprinting and tissue engineering.


Assuntos
Bioimpressão , Tecidos Suporte , Tecidos Suporte/química , Alginatos/química , Engenharia Tecidual , Hidrogéis/química , Gelatina/química , Impressão Tridimensional
9.
Stem Cells Transl Med ; 11(11): 1165-1176, 2022 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-36222619

RESUMO

Heterotopic ossification (HO) is a dynamic, complex pathologic process that often occurs after severe polytrauma trauma, resulting in an abnormal mesenchymal stem cell differentiation leading to ectopic bone growth in soft-tissues including tendons, ligaments, and muscles. The abnormal bone structure and location induce pain and loss of mobility. Recently, we observed that NGF (Nerve growth factor)-responsive TrkA (Tropomyosin receptor kinase A)-expressing nerves invade sites of soft-tissue trauma, and this is a necessary feature for heterotopic bone formation at sites of injury. Here, we assayed the effects of the partial TrkA agonist Gambogic amide (GA) in peritendinous heterotopic bone after extremity trauma. Mice underwent HO induction using the burn/tenotomy model with or without systemic treatment with GA, followed by an examination of the injury site via radiographic imaging, histology, and immunohistochemistry. Single-cell RNA Sequencing confirmed an increase in neurotrophin signaling activity after HO-inducing extremity trauma. Next, TrkA agonism led to injury site hyper-innervation, more brisk expression of cartilage antigens within the injured tendon, and a shift from FGF to TGFß signaling activity among injury site cells. Nine weeks after injury, this culminated in higher overall levels of heterotopic bone among GA-treated animals. In summary, these studies further link injury site hyper-innervation with increased vascular ingrowth and ultimately heterotopic bone after trauma. In the future, modulation of TrkA signaling may represent a potent means to prevent the trauma-induced heterotopic bone formation and improve tissue regeneration.


Assuntos
Queimaduras , Ossificação Heterotópica , Camundongos , Animais , Modelos Animais de Doenças , Ossificação Heterotópica/patologia , Tenotomia , Neurônios/patologia , Osteogênese
10.
Acta Biomater ; 154: 108-122, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36272687

RESUMO

Biological and mechanical cues are both vital for biomaterial aided tendon repair and regeneration. Here, we fabricated mechanically tendon-like (0 s UV) QHM polyurethane scaffolds (Q: Quadrol, H: Hexamethylene diisocyanate; M: Methacrylic anhydride) and immobilized them with Growth and differentiation factor-7 (GDF-7) to produce mechanically strong and tenogenic scaffolds. In this study, we assessed QHM polymer cytocompatibility, amenability to fibrin-coating, immobilization and persistence of GDF-7, and capability to support GDF-7-mediated tendon differentiation in vitro as well as in vivo in mouse subcutaneous and acute rat rotator cuff tendon resection models. Cytocompatibility studies showed that QHM facilitated cell attachment, proliferation, and viability. Fibrin-coating and GDF-7 retention studies showed that mechanically tendon-like 0 s UV QHM polymer could be immobilized with GDF-7 and retained the growth factor (GF) for at least 1-week ex vivo. In vitro differentiation studies showed that GDF-7 mediated bone marrow-derived human mesenchymal stem cell (hMSC) tendon-like differentiation on 0 s UV QHM. Subcutaneous implantation of GDF-7-immobilized, fibrin-coated, QHM polymer in mice for 2 weeks demonstrated de novo formation of tendon-like tissue while implantation of GDF-7-immobilized, fibrin-coated, QHM polymer in a rat acute rotator cuff resection injury model indicated tendon-like tissue formation in situ and the absence of heterotopic ossification. Together, our work demonstrates a promising synthetic scaffold with human tendon-like biomechanical attributes as well as immobilized tenogenic GDF-7 for tendon repair and regeneration. STATEMENT OF SIGNIFICANCE: Biological activity and mechanical robustness are key features required for tendon-promoting biomaterials. While synthetic biomaterials can be mechanically robust, they often lack bioactivity. To biologically augment synthetic biomaterials, numerous drug and GF delivery strategies exist but the large tissue space within the shoulder is constantly flushed with saline during arthroscopic surgery, hindering efficacious controlled release of therapeutic molecules. Here, we coated QHM polymer (which exhibits human tendon-to-bone-like biomechanical attributes) with fibrin for GF binding. Unlike conventional drug delivery strategies, our approach utilizes immobilized GFs as opposed to released GFs for sustained, localized tissue regeneration. Our data demonstrated that GF immobilization can be broadly applied to synthetic biomaterials for enhancing bioactivity, and GDF-7-immobilized QHM exhibit high clinical translational potential for tendon repair.


Assuntos
Polímeros , Lesões do Manguito Rotador , Ratos , Camundongos , Humanos , Animais , Poliuretanos/farmacologia , Anidridos , Tendões , Diferenciação Celular , Materiais Biocompatíveis , Lesões do Manguito Rotador/cirurgia , Tecidos Suporte/química
11.
iScience ; 25(5): 104229, 2022 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-35494239

RESUMO

This review presents bioprinting methods, biomaterials, and printing strategies that may be used for composite tissue constructs for musculoskeletal applications. The printing methods discussed include those that are suitable for acellular and cellular components, and the biomaterials include soft and rigid components that are suitable for soft and/or hard tissues. We also present strategies that focus on the integration of cell-laden soft and acellular rigid components under a single printing platform. Given the structural and functional complexity of native musculoskeletal tissue, we envision that hybrid bioprinting, referred to as hybprinting, could provide unprecedented potential by combining different materials and bioprinting techniques to engineer and assemble modular tissues.

12.
Tissue Eng Part A ; 28(17-18): 760-769, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35357948

RESUMO

Critical-sized cranial bone defect remains a great clinical challenge. With advantages in regenerative medicine, injectable hydrogels incorporated with bioactive molecules show great potential in promoting cranial bone repair. Recently, we developed a dual delivery system by sequential release of bone morphogenetic protein 2 (BMP2) followed by insulin-like growth factor 1 (IGF1) in microparticles (MPs), and an injectable alginate/collagen (alg/col)-based hydrogel. In this study, we aim to evaluate the effect of dual delivery of BMP2 and IGF1 in MPs through the injectable hydrogel in critical-sized cranial bone defect healing. The gelatin MPs loaded with BMP2 and poly(lactic-co-glycolic acid)-poly(ethylene glycol)-carboxyl (PLGA-PEG-COOH) MPs loaded with IGF1 were prepared, respectively. The encapsulation efficiency and release profile of growth factors in MPs were measured. A cranial defect model was applied to evaluate the efficacy of the dual delivery system in bone regeneration. Adult Sprague Dawley rats were subjected to osteotomy to make an ⌀8-mm cranial defect. The injectable hydrogel containing MPs loaded with BMP2 (2 µg), IGF1 (2 µg), or a combination of BMP2 (1 µg) and IGF1 (1 µg) were injected to the defect site. New bone formation was evaluated by microcomputed tomography, histological analysis, and immunohistochemistry after 4 or 8 weeks. Data showed that dual delivery of the low-dose BMP2 and IGF1 in MPs through alg/col-based hydrogel successfully restored cranial bone as early as 4 weeks after implantation, whose effect was comparable to the single delivery of high-dose BMP2 in MPs. In conclusion, this study suggests that dual delivery of BMP2 and IGF1 in MPs in alg/col-based hydrogel achieves early bone regeneration in critical-sized bone defect, with advantage in reducing the dose of BMP2. Impact Statement Sequential release of bone morphogenetic protein 2 (BMP2) followed by insulin-like growth factor 1 (IGF1) in two different microparticles promotes critical-sized bone defect healing. This dual delivery system reduces the dose of BMP2 by supplementing IGF1, which may diminish the potential side effects of BMP2.


Assuntos
Proteína Morfogenética Óssea 2 , Hidrogéis , Alginatos/farmacologia , Animais , Proteína Morfogenética Óssea 2/farmacologia , Regeneração Óssea , Hidrogéis/química , Hidrogéis/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Ratos , Ratos Sprague-Dawley , Crânio/patologia , Microtomografia por Raio-X
13.
Injury ; 53(4): 1368-1374, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35078617

RESUMO

OBJECTIVES: High energy long bone fractures with critical bone loss are at risk for nonunion without strategic intervention. We hypothesize that a synthetic membrane implanted at a single stage improves bone healing in a preclinical nonunion model. METHODS: Using standard laboratory techniques, microspheres encapsulating bone morphogenic protein-2 (BMP2) or platelet derived growth factor (PDGF) were designed and coupled to a type 1 collagen sheet. Critical femoral defects were created in rats and stabilized by locked retrograde intramedullary nailing. The negative control group had an empty defect. The induced membrane group (positive control) had a polymethylmethacrylate spacer inserted into the defect for four weeks and replaced with a bare polycaprolactone/beta-tricalcium phosphate (PCL/ß-TCP) scaffold at a second stage. For the experimental groups, a bioactive synthetic membrane embedded with BMP2, PDGF or both enveloped a PCL/ß-TCP scaffold was implanted in a single stage. Serial radiographs were taken at 1, 4, 8, and 12 weeks postoperatively from the definitive procedure and evaluated by two blinded observers using a previously described scoring system to judge union as primary outcome. RESULTS: All experimental groups demonstrated better union than the negative control (p = 0.01). The groups with BMP2 incorporated into the membrane demonstrated higher average union scores than the other groups (p = 0.01). The induced membrane group performed similarly to the PDGF group. Complete union was only demonstrated in groups with BMP2-eluting membranes. CONCLUSIONS: A synthetic membrane comprised of type 1 collagen embedded with controlled release BMP2 improved union of critical bone defects in a preclinical nonunion model.


Assuntos
Fosfatos de Cálcio , Fixação Intramedular de Fraturas , Animais , Fosfatos de Cálcio/farmacologia , Fêmur , Humanos , Polimetil Metacrilato , Ratos
14.
Bioengineering (Basel) ; 8(11)2021 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-34821731

RESUMO

The use of genetically modified (GM) mesenchymal stromal cells (MSCs) and preconditioned MSCs (pMSCs) may provide further opportunities to improve the outcome of core decompression (CD) for the treatment of early-stage osteonecrosis of the femoral head (ONFH). GM interleukin-4 (IL4) over-expressing MSCs (IL4-MSCs), platelet-derived growth factor (PDGF)-BB over-expressing MSCs (PDGF-BB-MSCs), and IL4-PDGF-BB co-over-expressing MSCs (IL4-PDGF-BB-MSCs) and their respective pMSCs were used in this in vitro study and compared with respect to cell proliferation and osteogenic differentiation. IL4-MSCs, PDGF-BB-MSCs, IL4-PDGF-BB-MSCs, and each pMSC treatment significantly increased cell proliferation compared to the MSC group alone. The percentage of Alizarin red-stained area in the IL4-MSC and IL4-pMSC groups was significantly lower than in the MSC group. However, the percentage of Alizarin red-stained area in the PDGF-BB-MSC group was significantly higher than in the MSC and PDGF-BB-pMSC groups. The percentage of Alizarin red-stained area in the IL4-PDGF-BB-pMSC was significantly higher than in the IL4-PDGF-BB-MSC group. There were no significant differences in the percentage of Alizarin red-stained area between the MSC and IL4-PDGF-BB-pMSC groups. The use of PDGF-BB-MSCs or IL4-PDGF-BB-pMSCs increased cell proliferation. Furthermore, PDGF-BB-MSCs promoted osteogenic differentiation. The addition of GM MSCs may provide a useful supplementary cell-based therapy to CD for treatment of ONFH.

15.
Stem Cell Res Ther ; 12(1): 503, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34526115

RESUMO

BACKGROUND: Approximately one third of patients undergoing core decompression (CD) for early-stage osteonecrosis of the femoral head (ONFH) experience progression of the disease, and subsequently require total hip arthroplasty (THA). Thus, identifying adjunctive treatments to optimize bone regeneration during CD is an unmet clinical need. Platelet-derived growth factor (PDGF)-BB plays a central role in cell growth and differentiation. The aim of this study was to characterize mesenchymal stromal cells (MSCs) that were genetically modified to overexpress PDGF-BB (PDGF-BB-MSCs) in vitro and evaluate their therapeutic effect when injected into the bone tunnel at the time of CD in an in vivo rabbit model of steroid-associated ONFH. METHODS: In vitro studies: Rabbit MSCs were transduced with a lentivirus vector carrying the human PDGF-BB gene under the control of either the cytomegalovirus (CMV) or phosphoglycerate (PGK) promoter. The proliferative rate, PDGF-BB expression level, and osteogenic differentiation capacity of unmodified MSCs, CMV-PDGF-BB-MSCs, and PGK-PDGF-BB-MSCs were assessed. In vivo studies: Twenty-four male New Zealand white rabbits received an intramuscular (IM) injection of methylprednisolone 20 mg/kg. Four weeks later, the rabbits were divided into four groups: the CD group, the hydrogel [HG, (a collagen-alginate mixture)] group, the MSC group, and the PGK-PDGF-BB-MSC group. Eight weeks later, the rabbits were sacrificed, their femurs were harvested, and microCT, mechanical testing, and histological analyses were performed. RESULTS: In vitro studies: PGK-PDGF-BB-MSCs proliferated more rapidly than unmodified MSCs (P < 0.001) and CMV-PDGF-BB-MSCs (P < 0.05) at days 3 and 7. CMV-PDGF-BB-MSCs demonstrated greater PDGF-BB expression than PGK-PDGF-BB-MSCs (P < 0.01). However, PGK-PDGF-BB-MSCs exhibited greater alkaline phosphatase staining at 14 days (P < 0.01), and osteogenic differentiation at 28 days (P = 0.07) than CMV-PDGF-BB-MSCs. In vivo: The PGK-PDGF-BB-MSC group had a trend towards greater bone mineral density (BMD) than the CD group (P = 0.074). The PGK-PDGF-BB-MSC group demonstrated significantly lower numbers of empty lacunae (P < 0.001), greater osteoclast density (P < 0.01), and greater angiogenesis (P < 0.01) than the other treatment groups. CONCLUSION: The use of PGK-PDGF-BB-MSCs as an adjunctive treatment with CD may reduce progression of osteonecrosis and enhance bone regeneration and angiogenesis in the treatment of early-stage ONFH.


Assuntos
Necrose da Cabeça do Fêmur , Células-Tronco Mesenquimais , Osteonecrose , Animais , Becaplermina , Descompressão , Cabeça do Fêmur , Necrose da Cabeça do Fêmur/induzido quimicamente , Necrose da Cabeça do Fêmur/genética , Necrose da Cabeça do Fêmur/terapia , Humanos , Masculino , Osteogênese , Coelhos , Esteroides
16.
Biomaterials ; 277: 121083, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34488121

RESUMO

Tendon injuries are one of the most common musculoskeletal disorders that cause considerable morbidity and significantly compromise the patients' quality of life. The innate limited regenerative capacity of tendon poses a substantial treating challenge for clinicians. MicroRNAs (miRNAs) are a family of small non-coding RNAs that play a vital role in orchestrating many biological processes through post-transcriptional regulation. Increasing evidence reveals that miRNA-based therapeutics may serve as an innovative strategy for the treatment of tendon pathologies. In this review, we briefly present miRNA biogenesis, the role of miRNAs in tendon cell biology and their involvement in tendon injuries, followed by a summary of current miRNA-based approaches in tendon tissue engineering with a special focus on attenuating post-injury fibrosis. Next, we discuss the advantages of miRNA-functionalized scaffolds in achieving sustained and localized miRNA administration to minimize off-target effects, and thus hoping to inspire the development of effective miRNA delivery platforms specifically for tendon tissue engineering. We envision that advancement in miRNA-based therapeutics will herald a new era of tendon tissue engineering and pave a way for clinical translation for the treatments of tendon disorders.


Assuntos
MicroRNAs , Traumatismos dos Tendões , Humanos , MicroRNAs/genética , Qualidade de Vida , Traumatismos dos Tendões/genética , Traumatismos dos Tendões/terapia , Tendões , Engenharia Tecidual
17.
Materials (Basel) ; 14(12)2021 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-34200817

RESUMO

The design of zirconia-based scaffolds using conventional techniques for bone-regeneration applications has been studied extensively. Similar to dental applications, the use of three-dimensional (3D) zirconia-based ceramics for bone tissue engineering (BTE) has recently attracted considerable attention because of their high mechanical strength and biocompatibility. However, techniques to fabricate zirconia-based scaffolds for bone regeneration are in a stage of infancy. Hence, the biological activities of zirconia-based ceramics for bone-regeneration applications have not been fully investigated, in contrast to the well-established calcium phosphate-based ceramics for bone-regeneration applications. This paper outlines recent research developments and challenges concerning numerous three-dimensional (3D) zirconia-based scaffolds and reviews the associated fundamental fabrication techniques, key 3D fabrication developments and practical encounters to identify the optimal 3D fabrication technique for obtaining 3D zirconia-based scaffolds suitable for real-world applications. This review mainly summarized the articles that focused on in vitro and in vivo studies along with the fundamental mechanical characterizations on the 3D zirconia-based scaffolds.

18.
Biomaterials ; 275: 120972, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34186237

RESUMO

Cell-based therapy for augmentation of core decompression (CD) using mesenchymal stromal cells (MSCs) is a promising treatment for early stage osteonecrosis of the femoral head (ONFH). Recently, the therapeutic potential for immunomodulation of osteogenesis using preconditioned (with pro-inflammatory cytokines) MSCs (pMSCs), or by the timely resolution of inflammation using MSCs that over-express anti-inflammatory cytokines has been described. Here, pMSCs exposed to tumor necrosis factor-alpha and lipopolysaccharide for 3 days accelerated osteogenic differentiation in vitro. Furthermore, injection of pMSCs encapsulated with injectable hydrogels into the bone tunnel facilitated angiogenesis and osteogenesis in the femoral head in vivo, using rabbit bone marrow-derived MSCs and a model of corticosteroid-associated ONFH in rabbits. In contrast, in vitro and in vivo studies demonstrated that genetically-modified MSCs that over-express IL4 (IL4-MSCs), established by using a lentiviral vector carrying the rabbit IL4 gene under the cytomegalovirus promoter, accelerated proliferation of MSCs and decreased the percentage of empty lacunae in the femoral head. Therefore, adjunctive cell-based therapy of CD using pMSCs and IL4-MSCs may hold promise to heal osteonecrotic lesions in the early stage ONFH. These interventions must be applied in a temporally sensitive fashion, without interfering with the mandatory acute inflammatory phase of bone healing.


Assuntos
Corticosteroides/efeitos adversos , Necrose da Cabeça do Fêmur , Células-Tronco Mesenquimais , Animais , Medula Óssea , Cabeça do Fêmur , Necrose da Cabeça do Fêmur/induzido quimicamente , Necrose da Cabeça do Fêmur/terapia , Interleucina-4 , Osteogênese , Coelhos
19.
Mater Sci Eng C Mater Biol Appl ; 123: 111950, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33812579

RESUMO

For the formation of new bone in critical-sized bone defects, bioactive scaffolds with an interconnected porous network are necessary. Herein, we fabricated three-dimensional (3D) porous hybrid zirconia scaffolds to promote hybrid functionality, i.e., excellent mechanical properties and bioactive performance. Specifically, the 3D printed scaffolds were subjected to Zn-HA/glass composite coating on glass-infiltrated zirconia (ZC). In addition, to pertain the extracellular matrix of bone, biopolymer (alginate/gelatine) was embedded in a developed 3D construct (ZB and ZCB). A zirconia-printed scaffold (Z) group served as a control. The structural and mechanical properties of the constructed scaffolds were studied using essential characterization techniques. Furthermore, the biological performance of the designed scaffolds was tested by a sequence of in vitro cell tests, including the attachment, proliferation, and osteogenic differentiation of dental pulp cells (DPCs). The ZC and ZCB scaffolds exhibited 20% higher compression strength than the zirconia (Z) scaffolds. More importantly, the ZC constructs exhibited superior cell-adhesion, distribution, and osteogenic differentiation ability due to the synergistic effects of the composite coating. In addition, the biopolymer-embedded scaffolds (ZB, ZCB) showed an excellent biological and mechanical performance. Thus, our results suggest that the Zn-HA/glass composite-coated glass-infiltrated zirconia (ZC, ZCB) scaffolds are a dynamic approach to designing bioactive 3D scaffolds for the load-bearing bone regeneration applications.


Assuntos
Osteogênese , Engenharia Tecidual , Regeneração Óssea , Porosidade , Tecidos Suporte , Zircônio
20.
J Orthop Translat ; 28: 90-99, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33816112

RESUMO

Background/Objective: Core decompression (CD) with scaffold and cell-based therapies is a promising strategy for providing both mechanical support and regeneration of the osteonecrotic area for early stage osteonecrosis of the femoral head (ONFH). We designed a new 3D printed porous functionally-graded scaffold (FGS) with a central channel to facilitate delivery of transplanted cells in a hydrogel to the osteonecrotic area. However, the optimal porous structural design for the FGS for the engineering of bone in ONFH has not been elucidated. The aim of this study was to fabricate and evaluate two different porous structures (30% or 60% porosity) of the FGSs in corticosteroid-associated ONFH in rabbits. METHODS: Two different FGSs with 30% or 60% porosity containing a 1-mm central channel were 3D printed using polycaprolactone and ß-tricalcium phosphate. The FGS was 3-mm diameter and 32-mm length and was composed of three segments: 1-mm in length for the non-porous proximal segment, 22-mm in length for the porous (30% versus 60%) middle segment, and 9-mm in length for the 15% porous distal segment. Eighteen male New Zealand White rabbits were given a single dose of 20 â€‹mg/kg methylprednisolone acetate intramuscularly. Four weeks later, rabbits were divided into three groups: the CD group, the 30% porosity FGS group, and the 60% porosity FGS group. In the CD group, a 3-mm diameter drill hole was created into the left femoral head. In the FGS groups, a 30% or 60% porosity implant was inserted into the bone tunnel. Eight weeks postoperatively, femurs were harvested and microCT, mechanical, and histological analyses were performed. RESULTS: The actual porosity and pore size of the middle segments were 26.4% â€‹± â€‹2.3% and 699 â€‹± â€‹56 â€‹µm in the 30% porosity FGS, and 56.0% â€‹± â€‹4.5% and 999 â€‹± â€‹71 â€‹µm in the 60% porosity FGS, respectively using microCT analysis. Bone ingrowth ratio in the 30% porosity FGS group was 73.9% â€‹± â€‹15.8%, which was significantly higher than 39.5% â€‹± â€‹13.0% in the CD group on microCT (p â€‹< â€‹0.05). Bone ingrowth ratio in the 60% porosity FGS group (61.3% â€‹± â€‹30.1%) showed no significant differences compared to the other two groups. The stiffness at the bone tunnel site in the 30% porosity FGS group was 582.4 â€‹± â€‹192.3 â€‹N/mm3, which was significantly higher than 338.7 â€‹± â€‹164.6 â€‹N/mm3 in the 60% porosity FGS group during push-out testing (p â€‹< â€‹0.05). Hematoxylin and eosin staining exhibited thick and mature trabecular bone around the porous FGS in the 30% porosity FGS group, whereas thinner, more immature trabecular bone was seen around the porous FGS in the 60% porosity FGS group. CONCLUSION: These findings indicate that the 30% porosity FGS may enhance bone regeneration and have superior biomechanical properties in the bone tunnel after CD in ONFH, compared to the 60% porosity FGS. TRANSLATION POTENTIAL STATEMENT: The translational potential of this article: This FGS implant holds promise for improving outcomes of CD for early stage ONFH.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...