Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Appl Sci (Basel) ; 12(23)2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36970107

RESUMO

Articular cartilage injuries are a common source of joint pain and dysfunction. As articular cartilage is avascular, it exhibits a poor intrinsic healing capacity for self-repair. Clinically, osteochondral grafts are used to surgically restore the articular surface following injury. A significant challenge remains with the repair properties at the graft-host tissue interface as proper integration is critical toward restoring normal load distribution across the joint. A key to addressing poor tissue integration may involve optimizing mobilization of fibroblast-like synoviocytes (FLS) that exhibit chondrogenic potential and are derived from the adjacent synovium, the specialized connective tissue membrane that envelops the diarthrodial joint. Synovium-derived cells have been directly implicated in the native repair response of articular cartilage. Electrotherapeutics hold potential as low-cost, low-risk, non-invasive adjunctive therapies for promoting cartilage healing via cell-mediated repair. Pulsed electromagnetic fields (PEMFs) and applied direct current (DC) electric fields (EFs) via galvanotaxis are two potential therapeutic strategies to promote cartilage repair by stimulating the migration of FLS within a wound or defect site. PEMF chambers were calibrated to recapitulate clinical standards (1.5 ± 0.2 mT, 75 Hz, 1.3 ms duration). PEMF stimulation promoted bovine FLS migration using a 2D in vitro scratch assay to assess the rate of wound closure following cruciform injury. Galvanotaxis DC EF stimulation assisted FLS migration within a collagen hydrogel matrix in order to promote cartilage repair. A novel tissue-scale bioreactor capable of applying DC EFs in sterile culture conditions to 3D constructs was designed in order to track the increased recruitment of synovial repair cells via galvanotaxis from intact bovine synovium explants to the site of a cartilage wound injury. PEMF stimulation further modulated FLS migration into the bovine cartilage defect region. Biochemical composition, histological analysis, and gene expression revealed elevated GAG and collagen levels following PEMF treatment, indicative of its pro-anabolic effect. Together, PEMF and galvanotaxis DC EF modulation are electrotherapeutic strategies with complementary repair properties. Both procedures may enable direct migration or selective homing of target cells to defect sites, thus augmenting natural repair processes for improving cartilage repair and healing.

2.
Tissue Eng Part A ; 27(21-22): 1411-1421, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33752445

RESUMO

Fibrosis of the knee is a common disorder resulting from an aberrant wound healing response and is characterized by extracellular matrix deposition, joint contraction, and scar tissue formation. The principal regulator of the fibrotic cascade is transforming growth factor beta-1 (TGF-ß1), a factor that induces rapid proliferation and differentiation of resident fibroblasts. In this study, we demonstrate successful inhibition of TGF-ß1-driven myofibroblastic differentiation in human fibroblast-like synoviocytes using a small molecule TGF-ß1 receptor inhibitor, SB-431542. We also demonstrate successful encapsulation of SB-431542 in poly(D,L-lactide-co-glycolide) (PLGA) as a potential prophylactic treatment for arthrofibrosis and characterize drug release and bioactivity in a three-dimensional collagen gel contraction assay. We assessed the effects of TGF-ß1 and SB-431542 on cell proliferation and viability in monolayer cultures. Opposing dose-dependent trends were observed in cell proliferation, which increased in TGF-ß1-treated cultures and decreased in SB-431542-treated cultures relative to control (p < 0.05). SB-431542 was not cytotoxic at the concentrations studied (0-50 µM) and inhibited TGF-ß1-induced collagen gel contraction in a dose-dependent manner. Specifically, TGF-ß1-treated gels contracted to 18% ± 1% of their initial surface area, while gels treated with TGF-ß1 and ≥10 µM SB-431542 showed no evidence of contraction (p < 0.0001). Upon removal of the compound, all gels contracted to control levels after 44 h in culture, necessitating sustained delivery for prolonged inhibition. To this end, SB-431542 was encapsulated in PLGA microspheres (SBMS) that had an average diameter of 87.5 ± 24 µm and a loading capacity of 4.3 µg SB-431542 per milligram of SBMS. Functional assessment of SBMS revealed sustained inhibition of TGF-ß1-induced gel contraction as well as hallmark features of myofibroblastic differentiation, including α-smooth muscle actin expression and connective tissue growth factor production. These results suggest that SB-431542 may be used to counter TGF-ß1-driven events in the fibrotic cascade in the knee cartilage. Impact statement Arthrofibrosis is the most prevalent comorbidity resulting from orthopedic procedures such as total knee arthroplasty that is characterized by excess deposition and accumulation of extracellular matrix. Despite its prevalence, treatments are generally palliative, and there is no effective prophylactic therapy. We report that the small molecule transforming growth factor beta-1 (TGF-ß1) receptor inhibitor, SB-431542, can inhibit the TGF-ß1-driven myofibroblastic differentiation of fibroblast-like synoviocytes. To provide sustained inhibition, we explored the use of SB-laden microspheres as a prophylactic therapy in a three-dimensional contraction model of fibrosis and propose that such therapies will have the potential to improve the standard of care for arthrofibrosis.


Assuntos
Fator de Crescimento Transformador beta , Benzamidas , Dioxóis , Humanos
3.
Biotechnol Bioeng ; 117(5): 1584-1596, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31985051

RESUMO

Articular cartilage injuries are a common source of joint pain and dysfunction. We hypothesized that pulsed electromagnetic fields (PEMFs) would improve growth and healing of tissue-engineered cartilage grafts in a direction-dependent manner. PEMF stimulation of engineered cartilage constructs was first evaluated in vitro using passaged adult canine chondrocytes embedded in an agarose hydrogel scaffold. PEMF coils oriented parallel to the articular surface induced superior repair stiffness compared to both perpendicular PEMF (p = .026) and control (p = .012). This was correlated with increased glycosaminoglycan deposition in both parallel and perpendicular PEMF orientations compared to control (p = .010 and .028, respectively). Following in vitro optimization, the potential clinical translation of PEMF was evaluated in a preliminary in vivo preclinical adult canine model. Engineered osteochondral constructs (∅ 6 mm × 6 mm thick, devitalized bone base) were cultured to maturity and implanted into focal defects created in the stifle (knee) joint. To assess expedited early repair, animals were assessed after a 3-month recovery period, with microfracture repairs serving as an additional clinical control. In vivo, PEMF led to a greater likelihood of normal chondrocyte (odds ratio [OR]: 2.5, p = .051) and proteoglycan (OR: 5.0, p = .013) histological scores in engineered constructs. Interestingly, engineered constructs outperformed microfracture in clinical scoring, regardless of PEMF treatment (p < .05). Overall, the studies provided evidence that PEMF stimulation enhanced engineered cartilage growth and repair, demonstrating a potential low-cost, low-risk, noninvasive treatment modality for expediting early cartilage repair.


Assuntos
Cartilagem Articular/efeitos da radiação , Campos Eletromagnéticos , Engenharia Tecidual/métodos , Cicatrização/efeitos dos fármacos , Animais , Cartilagem Articular/lesões , Células Cultivadas , Condrócitos/efeitos da radiação , Cães , Masculino , Joelho de Quadrúpedes/lesões
4.
Acta Biomater ; 102: 326-340, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31805408

RESUMO

Articular cartilage defects are a common source of joint pain and dysfunction. We hypothesized that sustained low-dose dexamethasone (DEX) delivery via an acellular osteochondral implant would have a dual pro-anabolic and anti-catabolic effect, both supporting the functional integrity of adjacent graft and host tissue while also attenuating inflammation caused by iatrogenic injury. An acellular agarose hydrogel carrier with embedded DEX-loaded poly(lactic-co-glycolic) acid (PLGA) microspheres (DLMS) was developed to provide sustained release for at least 99 days. The DLMS implant was first evaluated in an in vitro pro-inflammatory model of cartilage degradation. The implant was chondroprotective, as indicated by maintenance of Young's modulus (EY) (p = 0.92) and GAG content (p = 1.0) in the presence of interleukin-1ß insult. In a subsequent preliminary in vivo experiment, an osteochondral autograft transfer was performed using a pre-clinical canine model. DLMS implants were press-fit into the autograft donor site and compared to intra-articular DEX injection (INJ) or no DEX (CTL). Functional scores for DLMS animals returned to baseline (p = 0.39), whereas CTL and INJ remained significantly worse at 6 months (p < 0.05). DLMS knees were significantly more likely to have improved OARSI scores for proteoglycan, chondrocyte, and collagen pathology (p < 0.05). However, no significant improvements in synovial fluid cytokine content were observed. In conclusion, utilizing a targeted DLMS implant, we observed in vitro chondroprotection in the presence of IL-1-induced degradation and improved in vivo functional outcomes. These improved outcomes were correlated with superior histological scores but not necessarily a dampened inflammatory response, suggesting a primarily pro-anabolic effect. STATEMENT OF SIGNIFICANCE: Articular cartilage defects are a common source of joint pain and dysfunction. Effective treatment of these injuries may prevent the progression of osteoarthritis and reduce the need for total joint replacement. Dexamethasone, a potent glucocorticoid with concomitant anti-catabolic and pro-anabolic effects on cartilage, may serve as an adjuvant for a variety of repair strategies. Utilizing a dexamethasone-loaded osteochondral implant with controlled release characteristics, we demonstrated in vitro chondroprotection in the presence of IL-1-induced degradation and improved in vivo functional outcomes following osteochondral repair. These improved outcomes were correlated with superior histological cartilage scores and minimal-to-no comorbidity, which is a risk with high dose dexamethasone injections. Using this model of cartilage restoration, we have for the first time shown the application of targeted, low-dose dexamethasone for improved healing in a preclinical model of focal defect repair.


Assuntos
Anti-Inflamatórios/uso terapêutico , Dexametasona/uso terapêutico , Portadores de Fármacos/química , Microesferas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Sefarose/química , Animais , Autoenxertos/transplante , Transplante Ósseo , Cartilagem Articular/transplante , Bovinos , Preparações de Ação Retardada , Cães , Membro Posterior/cirurgia
5.
J Orthop Res ; 37(9): 1979-1987, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31062877

RESUMO

The synovium plays a key role in the development of osteoarthritis, as evidenced by pathological changes to the tissue observed in both early and late stages of the disease. One such change is the attachment of cartilage wear particles to the synovial intima. While this phenomenon has been well observed clinically, little is known of the biological effects that such particles have on resident cells in the synovium. The present work investigates the hypothesis that cartilage wear particles elicit a pro-inflammatory response in diseased and healthy human fibroblast-like synoviocytes, like that induced by key cytokines in osteoarthritis. Fibroblast-like synoviocytes from 15 osteoarthritic human donors and a subset of three non-osteoarthritic donors were exposed to cartilage wear particles, interleukin-1α or tumor necrosis factor-α for 6 days and analyzed for proliferation, matrix production, and release of pro-inflammatory mediators and degradative enzymes. Wear particles significantly increased proliferation and release of nitric oxide, interleukin-6 and -8, and matrix metalloproteinase-9, -10, and -13 in osteoarthritic synoviocytes, mirroring the effects of both cytokines, with similar trends in non-osteoarthritic cells. These results suggest that cartilage wear particles are a relevant physical factor in the osteoarthritic environment, perpetuating the pro-inflammatory and pro-degradative cascade by modulating synoviocyte behavior at early and late stages of the disease. Future work points to therapeutic strategies for slowing disease progression that target cell-particle interactions. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1979-1987, 2019.


Assuntos
Cartilagem/fisiologia , Citocinas/farmacologia , Inflamação/etiologia , Sinoviócitos/imunologia , Idoso , Idoso de 80 Anos ou mais , Feminino , Fibroblastos/imunologia , Humanos , Interleucina-1/farmacologia , Masculino , Pessoa de Meia-Idade , Osteoartrite/etiologia , Fator de Necrose Tumoral alfa/farmacologia
6.
Tissue Eng Part A ; 25(7-8): 538-553, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30203722

RESUMO

IMPACT STATEMENT: The synovium envelops the diarthrodial joint and plays a key regulatory role in defining the composition of the synovial fluid through filtration and biosynthesis of critical boundary lubricants. Synovium changes often precede cartilage damage in osteoarthritis. We describe a novel in vitro tissue engineered model, validated against native synovium explants, to investigate the structure-function of synovium through quantitative solute transport measures. Synovium was evaluated in the presence of a proinflammatory cytokine, interleukin-1, or the clinically relevant corticosteroid, dexamethasone. We anticipate that a better understanding of synovium transport would support efforts to develop more effective strategies aimed at restoring joint health.


Assuntos
Osteoartrite/metabolismo , Osteoartrite/patologia , Membrana Sinovial/citologia , Membrana Sinovial/metabolismo , Engenharia Tecidual/métodos , Corticosteroides/metabolismo , Cartilagem Articular/citologia , Cartilagem Articular/metabolismo , Citocinas/metabolismo , Dexametasona/metabolismo , Humanos , Interleucina-1/metabolismo , Líquido Sinovial/metabolismo
7.
J Orthop Res ; 34(1): 141-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26296185

RESUMO

With limited availability of osteochondral allografts, tissue engineered cartilage grafts may provide an alternative treatment for large cartilage defects. An effective storage protocol will be critical for translating this technology to clinical use. The purpose of this study was to evaluate the efficacy of the Missouri Osteochondral Allograft Preservation System (MOPS) for room temperature storage of mature tissue engineered grafts, focusing on tissue property maintenance during the current allograft storage window (28 days). Additional research compares MOPS to continued culture, investigates temperature influence, and examines longer-term storage. Articular cartilage constructs were cultured to maturity using adult canine chondrocytes, then preserved with MOPS at room temperature, in refrigeration, or kept in culture for an additional 56 days. MOPS storage maintained desired chondrocyte viability for 28 days of room temperature storage, retaining 75% of the maturity point Young's modulus without significant decline in biochemical content. Properties dropped past this time point. Refrigeration maintained properties similar to room temperature at 28 days, but proved better at 56 days. For engineered grafts, MOPS maintained the majority of tissue properties for the 28-day window without clearly extending that period as it had for native grafts. These results are the first evaluating engineered cartilage storage.


Assuntos
Cartilagem Articular , Engenharia Tecidual , Preservação de Tecido/métodos , Animais , Cães , Distribuição Aleatória
8.
Acta Biomater ; 8(5): 1891-900, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22326788

RESUMO

The strength and stability of hybrid fiber delivery systems, ones that perform a mechanical function and simultaneously deliver drug, are critical in the design of surgically implantable constructs. We report the fabrication of drug-eluting microfibers where drug loading and processing conditions alone increase microfiber strength and stability partially due to solvent-induced crystallization. Poly(L-lactic acid) microfibers of 64±7 µm diameter were wet spun by phase inversion. Encapsulation of a model hydrophobic anti-inflammatory drug, dexamethasone, at high loading provided stability to microfibers which maintained linear cumulative release kinetics up to 8 weeks in vitro. In our wet spinning process, all microfibers had increased crystallinity (13-17%) in comparison to unprocessed polymer without any mechanical stretching. Moreover, microfibers with the highest drug loading retained 97% of initial tensile strength and were statistically stronger than all other microfiber formulations, including control fibers without drug. Results indicate that the encapsulation of small hydrophobic molecules (<400 Da) may increase the mechanical integrity of microfilaments whose crystallinity is also increased as a result of the process. Multifunctional drug-eluting microfibers can provide an exciting new opportunity to design novel biomaterials with mechanical stability and controlled release of a variety of therapeutics with micron-scale accuracy.


Assuntos
Cápsulas/química , Preparações de Ação Retardada/química , Dexametasona/química , Ácido Láctico/química , Polímeros/química , Anti-Inflamatórios/química , Difusão , Teste de Materiais , Poliésteres
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...