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








Base de dados
Intervalo de ano de publicação
1.
Ann Biomed Eng ; 2023 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-36952145

RESUMO

Due to the limited self-repair capacity of the hyaline cartilage, the repair of cartilage remains an unsolved clinical problem. Tissue engineering strategy with 3D bioprinting technique has emerged a new insight by providing patient's personalized cartilage grafts using autologous cells for hyaline cartilage repair and regeneration. In this review, we first summarized the intrinsic property of hyaline cartilage in both maxillofacial and orthopedic regions to establish the requirement for 3D bioprinting cartilage tissue. We then reviewed the literature and provided opinion pieces on the selection of bioprinters, bioink materials, and cell sources. This review aims to identify the current challenges for hyaline cartilage bioprinting and the directions for future clinical development in bioprinted hyaline cartilage.

2.
Int J Mol Sci ; 23(13)2022 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-35805907

RESUMO

Nasal chondrocytes (NCs) have a higher and more reproducible chondrogenic capacity than articular chondrocytes, and the engineered cartilage tissue they generate in vitro has been demonstrated to be safe in clinical applications. Here, we aimed at determining the feasibility for a single-stage application of NCs for cartilage regeneration under minimally invasive settings. In particular, we assessed whether NCs isolated using a short collagenase digestion protocol retain their potential to proliferate and chondro-differentiate within an injectable, swiftly cross-linked and matrix-metalloproteinase (MMP)-degradable polyethylene glycol (PEG) gel enriched with human platelet lysate (hPL). NC-hPL-PEG gels were additionally tested for their capacity to generate cartilage tissue in vivo and to integrate into cartilage/bone compartments of human osteochondral plugs upon ectopic subcutaneous implantation into nude mice. NCs isolated with a rapid protocol and embedded in PEG gels with hPL at low cell density were capable of efficiently proliferating and of generating tissue rich in glycosaminoglycans and collagen II. NC-hPL-PEG gels developed into hyaline-like cartilage tissues upon ectopic in vivo implantation and integrated with surrounding native cartilage and bone tissues. The delivery of NCs in PEG gels containing hPL is a feasible strategy for cartilage repair and now requires further validation in orthotopic in vivo models.


Assuntos
Cartilagem Articular , Condrócitos , Animais , Humanos , Cartilagem Hialina , Hidrogéis , Camundongos , Camundongos Nus , Polietilenoglicóis/farmacologia , Engenharia Tecidual/métodos
3.
Cartilage ; 13(1): 19476035221075951, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35189712

RESUMO

OBJECTIVE: Implantation of tissue-engineered tracheal grafts represents a visionary strategy for the reconstruction of tracheal wall defects after resections and may develop into a last chance for a number of patients with severe cicatricial stenosis. The use of a decellularized tracheal substrate would offer an ideally stiff graft, but the matrix density would challenge efficient remodeling into a living cartilage. In this study, we hypothesized that the pores of decellularized laser-perforated tracheal cartilage (LPTC) tissues can be colonized by adult nasal chondrocytes (NCs) to produce new cartilage tissue suitable for the repair of tracheal defects. DESIGN: Human, native tracheal specimens, isolated from cadaveric donors, were exposed to decellularized and laser engraving-controlled superficial perforation (300 µm depth). Human or rabbit NCs were cultured on the LPTCs for 1 week. The resulting revitalized tissues were implanted ectopically in nude mice or orthotopically in tracheal wall defects in rabbits. Tissues were assayed histologically and by microtomography analyses before and after implantation. RESULTS: NCs were able to efficiently colonize the pores of the LPTCs. The extent of colonization (i.e., percentage of viable cells spanning >300 µm of tissue depth), cell morphology, and cartilage matrix deposition improved once the revitalized constructs were implanted ectopically in nude mice. LPTCs could be successfully grafted onto the tracheal wall of rabbits without any evidence of dislocation or tracheal stenosis, 8 weeks after implantation. Rabbit NCs, within the LPTCs, actively produced new cartilage matrix. CONCLUSION: Implantation of NC-revitalized LPTCs represents a feasible strategy for the repair of tracheal wall defects.


Assuntos
Gravuras e Gravação , Engenharia Tecidual , Animais , Cartilagem/transplante , Humanos , Lasers , Camundongos , Camundongos Nus , Coelhos , Engenharia Tecidual/métodos , Alicerces Teciduais
4.
Cartilage ; 13(2_suppl): 521S-529S, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34541930

RESUMO

OBJECTIVE: Nasal septal pathologies requiring surgical intervention are common in the population. Additionally, nasal chondrocytes are becoming an important cell source in cartilage tissue engineering strategies for the repair of articular cartilage lesions. These procedures damage the nasal septal cartilage whose healing potential is limited due to its avascular, aneural, and alymphatic nature. Despite the high incidence of various surgical interventions that affect septum cartilage, limited nasal cartilage repair characterizations have been performed to date. METHODS: To evaluate the healing of the nasal septum cartilage perforation, a septal biopsy was performed in 14 sheep. Two and 6 months later, the tissue formed on the place of perforation was explanted and compared with the native tissue. Tissue morphology, protein and gene expression of explanted tissue was determined using histological, immunohistochemical and real-time quantitative polymerase chain reaction analysis. RESULTS: Tissue formed on the defect site, 2 and 6 months after the biopsy was characterized as mostly connective tissue with the presence of fibroblastic cells. This newly formed tissue contained no glycosaminoglycans and collagen type II but was positively stained for collagen type I. Cartilage-specific genes COL2, AGG, and COMP were significantly decreased in 2- and 6-month samples compared with the native nasal cartilage. Levels of COL1, COL4, and CRABP1 genes specific for perichondrium and connective tissue were higher in both test group samples in comparison with native cartilage. CONCLUSIONS: Newly formed tissue was not cartilage but rather fibrous tissue suggesting the role of perichondrium and mucosa in tissue repair after nasal septum injury.


Assuntos
Cartilagem Articular , Condrócitos , Animais , Biópsia , Cartilagem Articular/patologia , Condrócitos/metabolismo , Septo Nasal , Ovinos , Engenharia Tecidual/métodos
5.
Am J Sports Med ; 49(8): 2187-2198, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34048271

RESUMO

BACKGROUND: Bipolar or "kissing" cartilage lesions formed on 2 opposite articular surfaces of the knee joint are commonly listed as exclusion criteria for advanced cartilage therapies. PURPOSE: To test, in a pilot large-animal study, whether autologous nasal chondrocyte (NC)-based tissue engineering, recently introduced for the treatment of focal cartilage injuries, could provide a solution for challenging kissing lesions. STUDY DESIGN: Controlled laboratory study. METHODS: Osteochondral kissing lesions were freshly introduced into the knee joints of 26 sheep and covered with NC-based grafts with a low or high hyaline-like extracellular matrix; a control group was treated with a cell-free scaffold collagen membrane (SCA). The cartilage repair site was assessed at 6 weeks and 6 months after implantation by histology, immunohistochemistry, and magnetic resonance imaging evaluation. RESULTS: NC-based grafts, independently of their composition, induced partial hyaline cartilage repair with stable integrity in surrounding healthy tissue at 6 months after treatment. The SCA repaired cartilage to a similar degree to that of NC-based grafts. CONCLUSION: Kissing lesion repair, as evidenced in this sheep study, demonstrated the feasibility of the treatment of complex cartilage injuries with advanced biological methods. However, the potential advantages of an NC-based approach over a cell-free approach warrant further investigations in a more relevant preclinical model. CLINICAL RELEVANCE: NC-based grafts currently undergoing phase II clinical trials have a high potential to replace existing cartilage therapies that show significant limitations in the quality and reproducibility of the repair method. We have brought this innovative concept to the next level by addressing a new clinical indication.


Assuntos
Cartilagem Articular , Animais , Cartilagem Articular/cirurgia , Condrócitos , Cartilagem Hialina , Articulação do Joelho , Reprodutibilidade dos Testes , Ovinos , Engenharia Tecidual , Transplante Autólogo
6.
Ann Biomed Eng ; 49(6): 1572-1586, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33409853

RESUMO

The treatment of cartilage defect remains a challenging issue in clinical practice. Chitosan-based materials have been recognized as a suitable microenvironment for chondrocyte adhesion, proliferation and differentiation forming articular cartilage. The use of nasal chondrocytes to culture articular cartilage on an appropriate scaffold emerged as a promising novel strategy for cartilage regeneration. Beside excellent properties, chitosan lacks in biological activity, such as RGD-sequences. In this work, we have prepared pure and protein-modified chitosan scaffolds of different deacetylation degree and molecular weight as platforms for the culture of sheep nasal chondrocytes. Fibronectin (FN) was chosen as an adhesive protein for the improvement of chitosan bioactivity. Prepared scaffolds were characterised in terms of microstructure, physical and biodegradation properties, while FN interactions with different chitosans were investigated through adsorption-desorption studies. The results indicated faster enzymatic degradation of chitosan scaffolds with lower deacetylation degree, while better FN interactions with material were achieved on chitosan with higher number of amine groups. Histological and immunohistochemical analysis of in vitro engineered cartilage grafts showed presence of hyaline cartilage produced by nasal chondrocytes.


Assuntos
Cartilagem Articular , Quitosana , Condrócitos/fisiologia , Alicerces Teciduais , Adsorção , Agrecanas/metabolismo , Animais , Cartilagem Articular/metabolismo , Adesão Celular , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Colágeno Tipo II/metabolismo , Fibronectinas/química , Septo Nasal/citologia , Fenazinas/metabolismo , Ovinos , Engenharia Tecidual/métodos
7.
Tissue Eng Part A ; 27(1-2): 37-49, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32122264

RESUMO

To investigate the effect of soluble factors released from human nasal chondrocytes (NCs) on cocultured human bone marrow mesenchymal stem cells (MSCs) and NC tissue-engineered constructs. Cartilage engineered from pure NCs on a three-dimensional (3D) porous collagen scaffold was cultured indirectly in a Transwell system with cartilage engineered from a direct coculture of human bone marrow-derived MSCs and NCs on a 3D porous collagen scaffold. The soluble factors were measured in the conditioned media from the different chambers of the Transwell system. Engineered cartilage from cocultures exposed to the pure NC construct exhibited reduced chondrogenic potential relative to control constructs, shown by reduced extracellular matrix deposition and increased expression of hypertrophic markers. Analysis of the soluble factors within the conditioned media showed an increase in inflammatory cytokines in the coculture chamber exposed to the pure NC construct. Principal component analysis revealed that the majority of the data variance could be explained by proinflammatory factors and hypertrophic chondrogenesis. In conclusion, our data suggest that inflammatory cytokines derived from NCs reduce the chondrogenic potential of coculture engineered cartilage through the induction of hypertrophic chondrogenesis. Impact statement The use of engineered cartilage from cocultured nasal chondrocytes (NCs) and mesenchymal stem cells for nasal cartilage reconstruction may be problematic. Our data suggest that the soluble factors from surrounding native NCs in the cartilage to be fixed can compromise the quality of the engineered cartilage if used in reconstructive surgery.


Assuntos
Condrogênese , Células-Tronco Mesenquimais , Diferenciação Celular , Células Cultivadas , Condrócitos , Técnicas de Cocultura , Humanos , Cartilagens Nasais , Engenharia Tecidual
8.
Bioengineering (Basel) ; 7(4)2020 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-33171883

RESUMO

One of the challenges in 3D-bioprinting is the realization of complex, volumetrically defined structures, that are also anatomically accurate and relevant. Towards this end, in this study we report the development and validation of a carboxylated agarose (CA)-based bioink that is amenable to 3D printing of free-standing structures with high stiffness at physiological temperature using microextrusion printing without the need for a fugitive phase or post-processing or support material (FRESH). By blending CA with negligible amounts of native agarose (NA) a bioink formulation (CANA) which is suitable for printing with nozzles of varying internal diameters under ideal pneumatic pressure was developed. The ability of the CANA ink to exhibit reproducible sol-gel transition at physiological temperature of 37 °C was established through rigorous characterization of the thermal behavior, and rheological properties. Using a customized bioprinter equipped with temperature-controlled nozzle and print bed, high-aspect ratio objects possessing anatomically-relevant curvature and architecture have been printed with high print reproducibility and dimension fidelity. Objects printed with CANA bioink were found to be structurally stable over a wide temperature range of 4 °C to 37 °C, and exhibited robust layer-to-layer bonding and integration, with evenly stratified structures, and a porous interior that is conducive to fluid transport. This exceptional layer-to-layer fusion (bonding) afforded by the CANA bioink during the print obviated the need for post-processing to stabilize printed structures. As a result, this novel CANA bioink is capable of yielding large (5-10 mm tall) free-standing objects ranging from simple tall cylinders, hemispheres, bifurcated 'Y'-shaped and 'S'-shaped hollow tubes, and cylinders with compartments without the need for support and/or a fugitive phase. Studies with human nasal chondrocytes showed that the CANA bioink is amenable to the incorporation of high density of cells (30 million/mL) without impact on printability. Furthermore, printed cells showed high viability and underwent mitosis which is necessary for promoting remodeling processes. The ability to print complex structures with high cell densities, combined with excellent cell and tissue biocompatibility of CA bodes well for the exploitation of CANA bioinks as a versatile 3D-bioprinting platform for the clinical translation of regenerative paradigms.

9.
Cell Mol Life Sci ; 77(23): 4847-4859, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32504256

RESUMO

Due to poor self-regenerative potential of articular cartilage, stem cell-based regeneration becomes a hopeful approach for the treatment of articular cartilage defects. Recent studies indicate that neural crest-derived cells (NCDCs) have the potential for repairing articular cartilage with even greater chondrogenic capacity than mesoderm-derived cells (MDCs): a conventional stem cell source for cartilage regeneration. Given that NCDCs originate from a different germ layer in the early embryo compared with MDCs that give rise to articular cartilage, a mystery remains regarding their capacity for articular cartilage regeneration. In this review, we summarize the similarities and differences between MDCs and NCDCs including articular and nasal chondrocytes in cell origin, anatomy, and chondrogenic differentiation and propose that NCDCs might be promising cell origins for articular cartilage regeneration.


Assuntos
Cartilagem Articular/fisiologia , Condrócitos/citologia , Crista Neural/citologia , Nariz/citologia , Regeneração , Células-Tronco/citologia , Animais , Humanos
10.
Cells ; 9(4)2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32295228

RESUMO

Chondrocyte transplantation has been successfully tested and proposed as a clinical procedure aiming to repair articular cartilage defects. However, the isolation of chondrocytes and the optimization of the enzymatic digestion process, as well as their successful in vitro expansion, remain the main challenges in cartilage tissue engineering. In order to address these issues, we investigated the performance of recombinant collagenases in tissue dissociation assays with the aim of isolating chondrocytes from bovine nasal cartilage in order to establish the optimal enzyme blend to ensure the best outcomes of the overall procedure. We show, for the first time, that collagenase H activity alone is required for effective cartilage digestion, resulting in an improvement in the yield of viable cells. The extracted chondrocytes proved able to grow and activate differentiation/dedifferentiation programs, as assessed by morphological and gene expression analyses.


Assuntos
Condrócitos/metabolismo , Crista Neural/metabolismo , Medicina Regenerativa/métodos , Engenharia Tecidual/métodos , Animais , Bovinos , Diferenciação Celular , Condrócitos/citologia , Humanos
11.
Clin Hemorheol Microcirc ; 74(1): 67-78, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31743993

RESUMO

OBJECTIVE: Implantation of autologous chondrocytes for cartilage repair requires harvesting of undamaged cartilage, implying an additional joint arthroscopy surgery and further damage to the articular surface. As alternative possible cell sources, in this study we assessed the proliferation and chondrogenic capacity of debrided Knee Chondrocytes (dKC) and Nasal Chondrocytes (NC) collected from the same patients. METHODS: Matched NC and dKC pairs from 13 patients enrolled in two clinical studies (NCT01605201 and NCT026739059) were expanded in monolayer and then chondro-differentiated in 3D collagenous scaffolds in medium with or without Transforming Growth Factor beta 1 (TGFß1). Cell proliferation and amount of cartilage matrix production by these two cell types were assessed. RESULTS: dKC exhibited an inferior proliferation rate than NC, and a lower capacity to chondro-differentiate. Resulting dKC-grafts contained lower amounts of cartilage specific matrix components glycosaminoglycans and type II collagen. The cartilage forming capacity of dKC did not significantly correlate with specific clinical parameters and was only partially improved by medium supplemention with TGFß1. CONCLUSIONS: dKC exhibit a reproducibly poor capacity to engineer cartilage grafts. Our in vitro data suggest that NC would be a better suitable cell source for the generation of autologous cartilage grafts.


Assuntos
Cartilagem Articular/fisiopatologia , Condrócitos/metabolismo , Articulação do Joelho/fisiopatologia , Nariz/fisiopatologia , Engenharia Tecidual/métodos , Adulto , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
12.
Front Bioeng Biotechnol ; 8: 572356, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33469528

RESUMO

OBJECTIVE: Human nasal septal chondrocytes (NC) are a promising minimally invasive derivable chondrogenic cell source for cartilage repair. However, the quality of NC-derived cartilage is variable between donors. Coculture of NC with mesenchymal stem cells (MSCs) mitigates the variability but with undesirable markers of chondrocyte hypertrophy, such as type X collagen, and the formation of unstable calcifying cartilage at ectopic sites. In contrast, monoculture NC forms non-calcifying stable cartilage. Formation of a stable NC-MSC coculture cartilage is crucial for clinical application. The aim of this study was to explore the utility of parathyroid hormone-related peptide (PTHrP) hormone to suppress chondrocyte hypertrophy in NC-MSC cocultures and form stable non-calcifying cartilage at ectopic sites. METHODS: Human NC and bone marrow MSCs, and cocultures of NC and MSC (1:3 ratio) were aggregated in pellet form and subjected to in vitro chondrogenesis for 3 weeks in chondrogenic medium in the presence and absence of PTHrP. Following in vitro chondrogenesis, the resulting pellets were implanted in immunodeficient athymic nude mice for 3 weeks. RESULTS: Coculture of NC and MSC resulted in synergistic cartilage matrix production. PTHrP suppressed the expression of hypertrophy marker, type X collagen (COL10A1), in a dose-dependent fashion without affecting the synergism in cartilage matrix synthesis, and in vivo calcification was eradicated with PTHrP. In contrast, cocultured control (CC) pellets without PTHrP treatment expressed COL10A1, calcified, and became vascularized in vivo.

13.
J Tissue Eng Regen Med ; 12(6): 1402-1411, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29726103

RESUMO

Bioreactor systems will likely play a key role in establishing regulatory compliant and cost-effective production systems for manufacturing engineered tissue grafts for clinical applications. However, the automation of bioreactor systems could become considerably more complex and costly due to the requirements for additional storage and liquid handling technologies if unstable supplements are added to the culture medium. Ascorbic acid (AA) is a bioactive supplement that is commonly presumed to be essential for the generation of engineered cartilage tissues. However, AA can be rapidly oxidized and degraded. In this work, we addressed whether human nasal chondrocytes can redifferentiate, undergo chondrogenesis, and generate a cartilaginous extracellular matrix when cultured in the absence of AA. We found that when chondrocytes were cultured in 3D micromass pellets either with or without AA, there were no significant differences in their chondrogenic capacity in terms of gene expression or the amount of glycosaminoglycans. Moreover, 3D pellets cultured without AA contained abundant collagen Types II and I extracellular matrix. Although the amounts of Collagens II and I were significantly lower (34% and 50% lower) than in pellets cultured with AA, collagen fibers had similar thicknesses and distributions for both groups, as shown by scanning electron microscopy imaging. Despite the reduced amounts of collagen, if engineered cartilage grafts can be generated with sufficient properties that meet defined quality criteria without the use of unstable supplements such as AA, bioreactor automation requirements can be greatly simplified, thereby facilitating the development of more compact, user-friendly, and cost-effective bioreactor-based manufacturing systems.


Assuntos
Ácido Ascórbico/farmacologia , Diferenciação Celular/efeitos dos fármacos , Condrócitos/citologia , Condrogênese , Adulto , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Condrogênese/genética , Colágeno/metabolismo , Meios de Cultura , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glicosaminoglicanos/metabolismo , Humanos , Pessoa de Meia-Idade , Adulto Jovem
14.
Biomaterials ; 171: 219-229, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29705655

RESUMO

Despite the various reported approaches to generate osteochondral composites by combination of different cell types and materials, engineering of templates with the capacity to autonomously and orderly develop into cartilage-bone bi-layered structures remains an open challenge. Here, we hypothesized that the embedding of cells inducible to endochondral ossification (i.e. bone marrow derived mesenchymal stromal cells, BMSCs) and of cells capable of robust and stable chondrogenesis (i.e. nasal chondrocytes, NCs) adjacent to each other in bi-layered hydrogels would develop directly in vivo into osteochondral tissues. Poly(ethylene glycol) (PEG) hydrogels were functionalized with TGFß3 or BMP-2, enzymatically polymerized encapsulating human BMSCs, combined with a hydrogel layer containing human NCs and ectopically implanted in nude mice without pre-culture. The BMSC-loaded layers reproducibly underwent endochondral ossification and generated ossicles containing bone and marrow. The NC-loaded layers formed cartilage tissues, which (under the influence of BMP-2 but not of TGFß3 from the neighbouring layer) remained phenotypically stable. The proposed strategy, resulting in orderly connected osteochondral composites, should be further assessed for the repair of osteoarticular defects and will be useful to model developmental processes leading to cartilage-bone interfaces.


Assuntos
Hidrogéis/farmacologia , Osteogênese/efeitos dos fármacos , Engenharia Tecidual/métodos , Adulto , Proteína Morfogenética Óssea 2/farmacologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Feminino , Humanos , Cartilagem Hialina/efeitos dos fármacos , Cartilagem Hialina/fisiologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Nariz/citologia , Polietilenoglicóis/farmacologia , Implantação de Prótese , Fator de Crescimento Transformador beta3/farmacologia
15.
Biomater Res ; 21: 19, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29075508

RESUMO

BACKGROUND: The three-dimensional (3D) system is one of the important factors to engineer a biocompatible and functional scaffold for the applications of cell-based therapies for cartilage repair. The 3D alginate hydrogels system has previously been shown to potentially promote chondrogenesis. The chondrocytic differentiation of co-cultured adipose-derived stem cells (ADSCs) and nasal chondrocytes (NCs) within alginate constructs are hypothesized to be influenced by concentration of alginate hydrogel. In this study, we evaluated the effects of alginate concentration on chondrogenic differentiation of ADSCs and NCs co-cultured in a biological approach. METHOD: The co-cultured cells of 2:1 ADSCs-to-NCs ratio were encapsulated in alginate constructs in one of three concentrations (1.0%, 1.2% and 1.5%) and cultured under serum free conditions for 7 days. Cell viability, cell proliferation, immunohistochemical, gycosaminogylycans (GAG) synthesis, and gene expression were examined. RESULTS: Overall, the 1.2% alginate concentration group was relatively effective in chondrocytic differentiation in comparable to other groups. The cell morphology, cell viability, and cell proliferation revealed initial chondrogenic differentiation by the formation of cell clusters as well as the high permeability for exchange of solutes. The formation of newly synthesis cartilage-specific extracellular matrix in 1.2% group was demonstrated by positive immunohistochemical staining of collagen type II. The co-cultured cells in 1.2% group highly expressed COL II, ACP and SOX-9, compared to 1.0% and 1.5% groups, denote the retention of cartilaginous-specific phenotype by suppressing the undifferentiation stem cell markers of SOX-2 and OCT-4. The study showed 1.2% group was less likely to differentiate towards osteogenesis by downregulating hyperthrophy chondrocytic gene of COL X and osseous marker genes of OSC and OSP. CONCLUSION: This study suggests that variations in the alginate concentration of co-cultured ADSCs and NCs influenced the chondrogenesis. The remarkable biological performance on chondrogenic differentiation in regulating the concentration of alginate 3D culture provides new insights into the cell cross-talk and demonstrates the effectiveness in regenerative therapies of cartilage defects in tissue engineering.

16.
J Tissue Eng Regen Med ; 11(3): 637-648, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-25370800

RESUMO

Limitations of current treatments for intervertebral disc (IVD) degeneration have promoted interest in the development of tissue-engineering approaches. Injectable hydrogels loaded with cells can be used as a substitute material for the inner IVD part, the nucleus pulposus (NP), and provide an opportunity for minimally invasive treatment of IVD degeneration. The NP is populated by chondrocyte-like cells; therefore, chondrocytes and mesenchymal stem cells (MSCs), stimulated to differentiate along the chondrogenic lineage, could be used to promote NP regeneration. In this study, the in vitro and in vivo response of human bone marrow-derived MSCs and nasal chondrocytes (NCs) to modified gellan gum-based hydrogels was investigated. Both ionic- (iGG-MA) and photo-crosslinked (phGG-MA) methacrylated gellan gum hydrogels show no cytotoxicity in extraction assays with MSCs and NCs. Furthermore, the materials do not induce pro-inflammatory responses in endothelial cells. Moreover, MSCs and NCs can be encapsulated into the hydrogels and remain viable for at least 2 weeks, although apoptosis is observed in phGG-MA. Importantly, encapsulated MSCs and NCs show signs of in vivo chondrogenesis in a subcutaneous implantation of iGG-MA. Altogether, the data endorse the potential use of modified gellan gum-based hydrogel as a suitable material in NP tissue engineering. Copyright © 2014 John Wiley & Sons, Ltd.


Assuntos
Células Imobilizadas/citologia , Hidrogéis/farmacologia , Metacrilatos/farmacologia , Núcleo Pulposo/fisiologia , Polissacarídeos Bacterianos/farmacologia , Regeneração/efeitos dos fármacos , Animais , Materiais Biocompatíveis/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Imobilizadas/efeitos dos fármacos , Células Imobilizadas/metabolismo , Condrogênese/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Marcação In Situ das Extremidades Cortadas , Inflamação/patologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/ultraestrutura , Camundongos SCID , Núcleo Pulposo/efeitos dos fármacos , Tela Subcutânea/efeitos dos fármacos
17.
Biomaterials ; 112: 313-323, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27770634

RESUMO

The regeneration of large bone defects remains clinically challenging. The aim of our study was to use a rat model to use nasal chondrocytes to engineer a hypertrophic cartilage tissue which could be remodelled into bone in vivo by endochondral ossification. Primary adult rat nasal chondrocytes were isolated from the nasal septum, the cell numbers expanded in monolayer culture and the cells cultured in vitro on polyglycolic acid scaffolds in chondrogenic medium for culture periods of 5-10 weeks. Hypertrophic differentiation was assessed by determining the temporal expression of key marker genes and proteins involved in hypertrophic cartilage formation. The temporal changes in the genes measured reflected the temporal changes observed in the growth plate. Collagen II gene expression increased 6 fold by day 7 and was then significantly downregulated from day 14 onwards. Conversely, collagen X gene expression was detectable by day 14 and increased 100-fold by day 35. The temporal increase in collagen X expression was mirrored by increases in alkaline phosphatase gene expression which also was detectable by day 14 with a 30-fold increase in gene expression by day 35. Histological and immunohistochemical analysis of the engineered constructs showed increased chondrocyte cell volume (31-45 µm), deposition of collagen X in the extracellular matrix and expression of alkaline phosphatase activity. However, no cartilage mineralisation was observed in in vitro culture of up to 10 weeks. On subcutaneous implantation of the hypertrophic engineered constructs, the grafts became vascularised, cartilage mineralisation occurred and loss of the proteoglycan in the matrix was observed. Implantation of the hypertrophic engineered constructs into a rat cranial defect resulted in angiogenesis, mineralisation and remodelling of the cartilage tissue into bone. Micro-CT analysis indicated that defects which received the engineered hypertrophic constructs showed 38.48% in bone volume compared to 7.01% in the control defects. Development of tissue engineered hypertrophic cartilage to use as a bone graft substitute is an exciting development in regenerative medicine. This is a proof of principal study demonstrating the potential of nasal chondrocytes to engineer hypertrophic cartilage which will remodel into bone on in vivo transplantation. This approach to making engineered hypertrophic cartilage grafts could form the basis of a new potential future clinical treatment for maxillofacial reconstruction.


Assuntos
Transplante Ósseo/instrumentação , Cartilagem/transplante , Condrócitos/transplante , Fraturas Cranianas/terapia , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Transplante Ósseo/métodos , Cartilagem/citologia , Cartilagem/crescimento & desenvolvimento , Células Cultivadas , Condrócitos/citologia , Condrócitos/fisiologia , Nariz/citologia , Ratos , Ratos Wistar , Fraturas Cranianas/patologia , Resultado do Tratamento
18.
J Cell Biochem ; 117(9): 2158-69, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27018169

RESUMO

Cartilage tissue engineering, with in vitro expansion of autologus chondrocytes, is a promising technique for tissue regeneration and is a new potential strategy to prevent and/or treat cartilage damage (e.g., osteoarthritis). The aim of this study was (i) to investigate and compare the effects of new biotechnological chondroitin (BC) and a commercial extractive chondroitin sulfate (CS) on human chondrocytes in vitro culture; (ii) to evaluate the anti-inflammatory effects of the innovative BC compared to extractive CS. A chondrogenic cell population was isolated from human nasoseptal cartilage and in vitro cultures were studied through time-lapse video microscopy (TLVM), immunohistochemical staining and cytometry. In order to investigate the effect of BC and CS on phenotype maintainance, chondrogenic gene expression of aggrecan (AGN), of the transcriptor factor SOX9, of the types I and II collagen (COL1A1 and COL1A2), were quantified through transcriptional and protein evaluation at increasing cultivation time and passages. In addition to resemble the osteoarthritis-like in vitro model, chondrocytes were treated with IL-1ß and the anti-inflammatory activity of BC and CS was assessed using cytokines quantification by multiplex array. BC significantly enhances cell proliferation also preserving chondrocyte phenotype increasing type II collagen expression up to 10 days of treatment and reduces inflammatory response in IL-1ß treated chondrocytes respect to CS treated cells. Our results, taken together, suggest that this new BC is of foremost importance in translational medicine because it can be applied in novel scaffolds and pharmaceutical preparations aiming at cartilage pathology treatments such as the osteoarthritis. J. Cell. Biochem. 117: 2158-2169, 2016. © 2016 The Authors. Journal of Cellular Biochemistry Published by Wiley Periodicals, Inc.


Assuntos
Antígenos de Diferenciação/biossíntese , Condrócitos/metabolismo , Condroitina/química , Regulação da Expressão Gênica , Alicerces Teciduais/química , Células Cultivadas , Condrócitos/citologia , Humanos
19.
Ann Transl Med ; 3(17): 254, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26605300

RESUMO

When a healthy joint progressively becomes osteoarthritic, the structures of the affected cartilage, bone and synovia undergo an initial phase of rearrangement. The exact molecular and cellular events occurring in this early osteoarthritic transition phase still remain elusive. Homeobox (Hox) genes encode for transcription factors that typically regulate limb morphogenesis and skeletal formation during development. More recently they were shown to be required for tissue remodelling and homeostasis in adults and to be modulated in a variety of pathologies. Here we present and discuss the hypothesis that dysregulation of specific Hox genes is associated with the onset and development of osteoarthritis (OA). Discovering mechanisms modulating Hox gene expression could not only provide important information in understanding OA pathology and its initiation, but also help to identify biomarkers reflecting the state of early OA. This knowledge would allow anticipating the time window for clinical treatment of the affected cartilage and assist in the development of innovative strategies to restore joint homeostasis, e.g., by cell or gene therapy.

20.
J Cell Mol Med ; 19(6): 1390-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25689393

RESUMO

Nasal chondrocytes (NC) derive from the same multipotent embryological segment that gives rise to the majority of the maxillofacial bone and have been reported to differentiate into osteoblast-like cells in vitro. In this study, we assessed the capacity of adult human NC, appropriately primed towards hypertrophic or osteoblastic differentiation, to form bone tissue in vivo. Hypertrophic induction of NC-based micromass pellets formed mineralized cartilaginous tissues rich in type X collagen, but upon implantation into subcutaneous pockets of nude mice remained avascular and reverted to stable hyaline-cartilage. In the same ectopic environment, NC embedded into ceramic scaffolds and primed with osteogenic medium only sporadically formed intramembranous bone tissue. A clonal study could not demonstrate that the low bone formation efficiency was related to a possibly small proportion of cells competent to become fully functional osteoblasts. We next tested whether the cues present in an orthotopic environment could induce a more efficient direct osteoblastic transformation of NC. Using a nude rat calvarial defect model, we demonstrated that (i) NC directly participated in frank bone formation and (ii) the efficiency of survival and bone formation by NC was significantly higher than that of reference osteogenic cells, namely bone marrow-derived mesenchymal stromal cells. This study provides a proof-of-principle that NC have the plasticity to convert into bone cells and thereby represent an easily available cell source to be further investigated for craniofacial bone regeneration.


Assuntos
Diferenciação Celular/fisiologia , Condrócitos/fisiologia , Septo Nasal/citologia , Osteoblastos/fisiologia , Osteogênese/fisiologia , Adulto , Idoso , Animais , Cartilagem/metabolismo , Cartilagem/fisiologia , Diferenciação Celular/genética , Células Cultivadas , Condrócitos/metabolismo , Feminino , Expressão Gênica , Humanos , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos Nus , Pessoa de Meia-Idade , Osteoblastos/metabolismo , Osteogênese/genética , Osteonectina/genética , Osteopontina/genética , Ratos Nus , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alicerces Teciduais , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA