Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cells Dev ; 29(14): 882-894, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32364057

RESUMO

Articular cartilage contains a subpopulation of tissue-specific progenitors that are an ideal cell type for cell therapies and generating neocartilage for tissue engineering applications. However, it is unclear whether the standard chondrogenic medium using transforming growth factor beta (TGFß) isoforms is optimal to differentiate these cells. We therefore used pellet culture to screen progenitors from immature bovine articular cartilage with a number of chondrogenic factors and discovered that bone morphogenetic protein-9 (BMP9) precociously induces their differentiation. This difference was apparent with toluidine blue staining and confirmed by biochemical and transcriptional analyses with BMP9-treated progenitors exhibiting 11-fold and 5-fold greater aggrecan and collagen type II (COL2A1) gene expression than TGFß1-treated progenitors. Quantitative gene expression analysis over 14 days highlighted the rapid and phased nature of BMP9-induced chondrogenesis with sequential activation of aggrecan then collagen type II, and negligible collagen type X gene expression. The extracellular matrix of TGFß1-treated progenitors analyzed using atomic force microscopy was fibrillar and stiff whist BMP9-induced matrix of cells more compliant and correspondingly less fibrillar. Polarized light microscopy revealed an annular pattern of collagen fibril deposition typified by TGFß1-treated pellets, whereas BMP9-treated pellets displayed a birefringence pattern that was more anisotropic. Remarkably, differentiated immature chondrocytes incubated as high-density cultures in vitro with BMP9 generated a pronounced anisotropic organization of collagen fibrils indistinguishable from mature adult articular cartilage, with cells in deeper zones arranged in columnar manner. This contrasted with cells grown with TGFß1, where a concentric pattern of collagen fibrils was visualized within tissue pellets. In summary, BMP9 is a potent chondrogenic factor for articular cartilage progenitors and is also capable of inducing morphogenesis of adult-like cartilage, a highly desirable attribute for in vitro tissue-engineered cartilage.


Assuntos
Cartilagem Articular/citologia , Condrogênese , Fator 2 de Diferenciação de Crescimento/metabolismo , Células-Tronco/citologia , Animais , Bovinos , Células Cultivadas , Colágeno/metabolismo , Regulação da Expressão Gênica , Fator 2 de Diferenciação de Crescimento/genética , Hidroxiprolina/metabolismo
2.
Int J Mol Sci ; 19(7)2018 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-29966376

RESUMO

The osteocyte network inside the bone matrix is of functional importance and osteocyte cell death is a characteristic feature of pathological bone diseases. Osteocytes have emerged as key regulators of bone tissue maintenance, yet maintaining their phenotype during in vitro culture remains challenging. A 3D co-culture system for osteocytes with osteoblasts was recently presented, enabling the determination of more physiological effects of growth factors on cells in vitro. MLO-Y4 cells were embedded within a type I collagen gel and cultured in the presence of surface MG-63 cells. Co-culture was performed in the presence or absence of TGFß3. Gene expression by quantitative PCR, protein expression by fluorescent immunohistochemistry and cell viability tests were performed. The 3D co-culture induced cell differentiation of MG-63 cells seen by increased type I collagen and osteocalcin mRNA expression. TGFβ3 maintained osteocyte differentiation of MLO-Y4 cells during co-culture as determined by stable E11 and osteocalcin mRNA expression till day 4. Interestingly, most of the effects of TGFß3 on co-cultured cells were serum-dependent. Also, TGFß3 reduced cell death of 3D co-cultured MLO-Y4 cells in a serum-dependent manner. This study shows that 3D co-culture upregulates differentiation of MG-63 cells to a more mature osteoblast-like phenotype; while the addition of TGFß3 maintained the characteristic MLO-Y4 osteocyte-like phenotype and viability in a serum-dependent manner.


Assuntos
Técnicas de Cocultura/métodos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Osteócitos/efeitos dos fármacos , Osteócitos/metabolismo
3.
Front Surg ; 4: 4, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28280722

RESUMO

Recent advances in microsurgery, imaging, and transplantation have led to significant refinements in autologous reconstructive options; however, the morbidity of donor sites remains. This would be eliminated by successful clinical translation of tissue-engineered solutions into surgical practice. Plastic surgeons are uniquely placed to be intrinsically involved in the research and development of laboratory engineered tissues and their subsequent use. In this article, we present an overview of the field of tissue engineering, with the practicing plastic surgeon in mind. The Medical Research Council states that regenerative medicine and tissue engineering "holds the promise of revolutionizing patient care in the twenty-first century." The UK government highlighted regenerative medicine as one of the key eight great technologies in their industrial strategy worthy of significant investment. The long-term aim of successful biomanufacture to repair composite defects depends on interdisciplinary collaboration between cell biologists, material scientists, engineers, and associated medical specialties; however currently, there is a current lack of coordination in the field as a whole. Barriers to translation are deep rooted at the basic science level, manifested by a lack of consensus on the ideal cell source, scaffold, molecular cues, and environment and manufacturing strategy. There is also insufficient understanding of the long-term safety and durability of tissue-engineered constructs. This review aims to highlight that individualized approaches to the field are not adequate, and research collaboratives will be essential to bring together differing areas of expertise to expedite future clinical translation. The use of tissue engineering in reconstructive surgery would result in a paradigm shift but it is important to maintain realistic expectations. It is generally accepted that it takes 20-30 years from the start of basic science research to clinical utility, demonstrated by contemporary treatments such as bone marrow transplantation. Although great advances have been made in the tissue engineering field, we highlight the barriers that need to be overcome before we see the routine use of tissue-engineered solutions.

4.
Sci Rep ; 7: 41421, 2017 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-28150695

RESUMO

In recent years it has become increasingly clear that articular cartilage harbours a viable pool of progenitor cells and interest has focussed on their role during development and disease. Analysis of progenitor numbers using fluorescence-activated sorting techniques has resulted in wide-ranging estimates, which may be the result of context-dependent expression of cell surface markers. We have used a colony-forming assay to reliably determine chondroprogenitor numbers in normal and osteoarthritic cartilage where we observed a 2-fold increase in diseased tissue (P < 0.0001). Intriguingly, cell kinetic analysis of clonal isolates derived from single and multiple donors of osteoarthritic cartilage revealed the presence of a divergent progenitor subpopulation characterised by an early senescent phenotype. Divergent sub-populations displayed increased senescence-associated ß-galactosidase activity, lower average telomere lengths but retained the capacity to undergo multi-lineage differentiation. Osteoarthritis is an age-related disease and cellular senescence is predicted to be a significant component of the pathological process. This study shows that although early senescence is an inherent property of a subset of activated progenitors, there is also a pool of progenitors with extended viability and regenerative potential residing within osteoarthritic cartilage.


Assuntos
Cartilagem Articular/patologia , Senescência Celular , Osteoartrite/patologia , Células-Tronco/patologia , Telômero/metabolismo , Adulto , Células-Tronco Adultas/patologia , Idoso , Idoso de 80 Anos ou mais , Bromodesoxiuridina/metabolismo , Núcleo Celular/metabolismo , Separação Celular , Cromossomos Humanos/metabolismo , Células Clonais , Humanos , Modelos Lineares , Pessoa de Meia-Idade , beta-Galactosidase/metabolismo
5.
J Tissue Eng Regen Med ; 11(10): 2912-2921, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-27406210

RESUMO

Mesenchymal stem cells (MSCs) are currently being investigated as candidate cells for regenerative medicine approaches for the repair of damaged articular cartilage. For these cells to be used clinically, it is important to understand how they will react to the complex loading environment of a joint in vivo. In addition to investigating alternative cell sources, it is also important for the structure of tissue-engineered constructs and the organization of cells within them to be developed and, if possible, improved. A custom built bioreactor was used to expose human MSCs to a combination of shear and compression loading. The MSCs were either evenly distributed throughout fibrin-poly(ester-urethane) scaffolds or asymmetrically seeded with a small proportion seeded on the surface of the scaffold. The effect of cell distribution on the production and deposition of cartilage-like matrix in response to mechanical load mimicking in vivo joint loading was then investigated. The results show that asymmetrically seeding the scaffold led to markedly improved tissue development based on histologically detectable matrix deposition. Consideration of cell location, therefore, is an important aspect in the development of regenerative medicine approaches for cartilage repair. This is particularly relevant when considering the natural biomechanical environment of the joint in vivo and patient rehabilitation protocols. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Condrogênese/efeitos dos fármacos , Fibrina/farmacologia , Células-Tronco Mesenquimais/citologia , Poliésteres/farmacologia , Poliuretanos/farmacologia , Alicerces Teciduais/química , Adolescente , Idoso , DNA/metabolismo , Feminino , Corantes Fluorescentes/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glicosaminoglicanos/metabolismo , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Coloração e Rotulagem , Adulto Jovem
6.
Stem Cell Res Ther ; 7: 19, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26822227

RESUMO

Recent advances in regenerative medicine place us in a unique position to improve the quality of engineered tissue. We use auricular cartilage as an exemplar to illustrate how the use of tissue-specific adult stem cells, assembly through additive manufacturing and improved understanding of postnatal tissue maturation will allow us to more accurately replicate native tissue anisotropy. This review highlights the limitations of autologous auricular reconstruction, including donor site morbidity, technical considerations and long-term complications. Current tissue-engineered auricular constructs implanted into immune-competent animal models have been observed to undergo inflammation, fibrosis, foreign body reaction, calcification and degradation. Combining biomimetic regenerative medicine strategies will allow us to improve tissue-engineered auricular cartilage with respect to biochemical composition and functionality, as well as microstructural organization and overall shape. Creating functional and durable tissue has the potential to shift the paradigm in reconstructive surgery by obviating the need for donor sites.


Assuntos
Cartilagem da Orelha/fisiologia , Animais , Pavilhão Auricular/fisiologia , Humanos , Especificidade de Órgãos , Procedimentos de Cirurgia Plástica , Regeneração , Medicina Regenerativa , Engenharia Tecidual
7.
PLoS One ; 10(8): e0136229, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26292283

RESUMO

Articular cartilage progenitor cells (ACPCs) represent a new and potentially powerful alternative cell source to commonly used cell sources for cartilage repair, such as chondrocytes and bone-marrow derived mesenchymal stem cells (MSCs). This is particularly due to the apparent resistance of ACPCs to hypertrophy. The current study opted to investigate whether human ACPCs (hACPCs) are responsive towards mechanical stimulation and/or adenoviral-mediated overexpression of bone morphogenetic protein 2 (BMP-2). hACPCs were cultured in fibrin-polyurethane composite scaffolds. Cells were cultured in a defined chondro-permissive medium, lacking exogenous growth factors. Constructs were cultured, for 7 or 28 days, under free-swelling conditions or with the application of complex mechanical stimulation, using a custom built bioreactor that is able to generate joint-like movements. Outcome parameters were quantification of BMP-2 and transforming growth factor beta 1 (TGF-ß1) concentration within the cell culture medium, biochemical and gene expression analyses, histology and immunohistochemistry. The application of mechanical stimulation alone resulted in the initiation of chondrogenesis, demonstrating the cells are mechanoresponsive. This was evidenced by increased GAG production, lack of expression of hypertrophic markers and a promising gene expression profile (significant up-regulation of cartilaginous marker genes, specifically collagen type II, accompanied by no increase in the hypertrophic marker collagen type X or the osteogenic marker alkaline phosphatase). To further investigate the resistance of ACPCs to hypertrophy, overexpression of a factor associated with hypertrophic differentiation, BMP-2, was investigated. A novel, three-dimensional, transduction protocol was used to transduce cells with an adenovirus coding for BMP-2. Over-expression of BMP-2, independent of load, led to an increase in markers associated with hypertropy. Taken together ACPCs represent a potential alterative cell source for cartilage tissue engineering applications.


Assuntos
Proteína Morfogenética Óssea 2/genética , Cartilagem Articular/citologia , Células-Tronco/citologia , Estresse Mecânico , Regulação para Cima , Adenoviridae/genética , Células Cultivadas , Condrogênese , Colágeno Tipo II/metabolismo , Fibrina/química , Regulação da Expressão Gênica , Vetores Genéticos/genética , Glicosaminoglicanos/metabolismo , Humanos , Células-Tronco/metabolismo , Alicerces Teciduais/química , Transdução Genética , Fator de Crescimento Transformador beta1/metabolismo
8.
J Bone Joint Surg Am ; 97(6): 484-93, 2015 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-25788305

RESUMO

BACKGROUND: We sought to determine the effectiveness of chondroprogenitor cells derived from autologous and allogenic articular cartilage for the repair of cartilage defects in an equine model. METHODS: Cartilage defects (15 mm) were created on the medial trochlear ridge of the femur. The following experimental treatments were compared with empty-defect controls: fibrin only, autologous chondroprogenitor cells plus fibrin, and allogenic chondroprogenitor cells plus fibrin (n = 4 or 12 per treatment). Horses underwent strenuous exercise throughout the twelve-month study, and evaluations included lameness (pain) and arthroscopic, radiographic, gross, histologic, and immunohistochemical analyses. RESULTS: Arthroscopy and microscopy indicated that defects in the autologous cell group had significantly better repair tissue compared with defects in the fibrin-only and control groups. Repair tissue quality in the allogenic cell group was not superior to that in the fibrin-only group with the exception of the percentage of type-II collagen, which was greater. Radiographic changes in the allogenic cell group were poorer on average than those in the autologous cell group. Autologous cells significantly reduced central osteophyte formation compared with fibrin alone. CONCLUSIONS: On the basis of the arthroscopic, radiographic, and histologic scores, autologous cells in fibrin yielded better results than the other treatments; allogenic cells cannot be recommended at this time.


Assuntos
Cartilagem Articular/lesões , Condrócitos/transplante , Transplante de Células-Tronco , Animais , Artroscopia , Cartilagem Articular/patologia , Modelos Animais de Doenças , Fêmur , Fibrina , Cavalos , Transplante Autólogo , Transplante Homólogo , Cicatrização
9.
Cartilage ; 5(4): 203-14, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26069699

RESUMO

OBJECTIVES: Osteoarthritis (OA) is a debilitating disease affecting more than 4 million people in the United Kingdom. Despite its prevalence, there is no successful cell-based therapy currently used to treat patients whose cartilage is deemed irrecoverable. The present study aimed to isolate stem cells from tibial plateaux cartilage obtained from patients who underwent total knee replacements for OA and investigate their stem cell characteristics. DESIGN: Clonally derived cell lines were selected using a differential adhesion assay to fibronectin and expanded in monolayer culture. Colony forming efficiencies and growth kinetics were investigated. The potential for tri-lineage differentiation into chondrogenic, osteogenic, and adipogenic phenotypes were analyzed using histological stains, immunocytochemistry, and reverse transcriptase polymerase chain reaction. RESULTS: Colony forming cells were successfully isolated from osteoarthritic cartilage and extensively expanded in monolayer culture. Colony forming efficiencies were consistently below 0.1%. Clonal cell lines were expanded beyond 40 population doublings but disparities were observed in the number of population doublings per day. Clonally derived cell lines also demonstrated in vitro multilineage potential via successful differentiation into chondrogenic, osteogenic, and adipogenic lineages. However, variation in the degree of differentiation was observed between these clonal cell lines. CONCLUSIONS: A viable pool of cells with stem cell characteristics have been identified within human osteoarthritic cartilage. Variation in the degree of differentiation suggests the possibility of further subpopulations of cells. The identification of this stem cell population highlights the reparative potential of these cells in osteoarthritic cartilage, which could be further exploited to aid the field of regenerative medicine.

10.
Cartilage ; 5(4): 231-40, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26069702

RESUMO

OBJECTIVE: Articular cartilage is a complex tissue comprising phenotypically distinct zones. Research has identified the presence of a progenitor cell population in the surface zone of immature articular cartilage. The aim of the present study was to determine the in vivo plasticity of articular cartilage progenitor. DESIGN: Chondropogenitor cells were isolated from bovine metacarpalphalangeal joints by differential adhesion to fibronectin. Cells were labeled with PKH26 and injected into the thigh muscle of severe-combined immunodeficient (SCID) mice. After 2 weeks, the muscles were dissected and cryosectioned. Sections were stained with safranin O and labeled for sox9 and collagen type II. Polymerase chain reaction analysis was carried out to determine plasticity for a number of tissue-specific markers. Full-depth chondrocytes acted as a control. RESULTS: Fluorescent PKH26 labeled cells were detected after 2 weeks in all samples analyzed. A cartilage pellet was present after injection of freshly isolated chondrocytes. After injection with clonal and enriched populations of chondroprogenitors, no distinct pellet was detected, but diffuse cartilage nodules were found with regions of safranin O staining and Sox9. Low levels of collagen type II were also detected. Polymerase chain reaction analysis identified the presence of the endothelial cell marker PECAM-1 in one clonal cell line, demonstrating phenotypic plasticity into the phenotype of the surrounding host tissues. CONCLUSIONS: The bovine articular cartilage progenitor cells were able to survive in vivo postimplantation, but failed to create a robust cartilage pellet, despite expressing sox9 and type II collagen. This suggests the cells require further signals for chondrogenic differentiation.

11.
Tissue Eng Part A ; 19(11-12): 1285-94, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23289669

RESUMO

Currently available methods to treat articular cartilage defects still fail to demonstrate satisfactory outcomes for many patients. Functional tissue engineering using human bone marrow-derived mesenchymal stem cells (hMSCs) is a promising alternative approach for the treatment of these defects. This study strived to investigate the combined effect of complex mechanical stimulation and adenoviral-mediated overexpression of bone morphogenetic protein 2 (BMP-2) on hMSC chondrogenesis. hMSCs were encapsulated in a fibrin hydrogel and seeded into biodegradable polyurethane (PU) scaffolds. A novel three-dimensional transduction protocol was used to transduce cells with an adenovirus encoding for BMP-2 (Ad.BMP-2). Control cells were left untransduced. Cells were cultured for 7 or 28 days in a chondropermessive medium, which lacks any exogenous growth factors. Thereby, the in vivo situation is mimicked more precisely. hMSCs in fibrin-PU composite scaffolds were either left as free-swelling controls or mechanically stimulated using a custom-built bioreactor system that is able to generate joint-like forces. Outcome parameters measured were BMP-2 concentration within the culture medium, and biochemical and gene expression analysis. Mechanical stimulation resulted in an upregulation of chondrogenic genes. Further, glycosaminoglycan (GAG)/DNA ratios were elevated in mechanically stimulated groups. Transduction with Ad.BMP-2 led to a pronounced upregulation of the gene aggrecan and an upregulation of Sox9 message after 7 days. Furthermore, a synergistic effect in combination with mechanical stimulation on collagen 2 message was detected after 7 days. This synergistic increase was more than 8-fold if compared to the additive effect of the application of each stimulus on its own. However, BMP-2 overexpression consistently resulted in a trend toward decreased GAG/DNA ratios in both mechanical stimulated and unloaded groups.


Assuntos
Adenoviridae/metabolismo , Células da Medula Óssea/citologia , Proteína Morfogenética Óssea 2/metabolismo , Condrogênese , Células-Tronco Mesenquimais/citologia , Estresse Mecânico , Agrecanas/genética , Agrecanas/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Condrogênese/efeitos dos fármacos , Condrogênese/genética , Meios de Cultura/farmacologia , DNA/metabolismo , Fibrina/farmacologia , Glicosaminoglicanos/metabolismo , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Poliuretanos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Transdução Genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
12.
Mol Biotechnol ; 53(2): 207-16, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22382454

RESUMO

Hydrogels are increasingly being investigated as a means to implant cells for tissue engineering. One way to further enhance the repair response would be to combine the hydrogel cell carrier with gene transfer. Gene therapy, using adenoviral vectors, is an effective way to provide transient delivery of bioactive factors. However, current protocols require further optimization, especially if they are to be transferred into the clinic. This study opted to compare the efficiency of protocols for standard two-dimensional (2D) versus three-dimensional (3D), adenoviral-mediated, transduction of human mesenchymal stem cells. Two different multiplicities of infection were tested. After encapsulation in fibrin, alginate or agarose, cells were cultured for 28 days. Transduction in 3D showed a much higher efficiency, compared to standard 2D transduction protocols. In 3D, the amount of transgene produced was significantly higher, for every condition investigated. Furthermore, transduction in 3D does not require a cell culture step and can be conducted within the operating theatre. In conclusion, it was demonstrated that 3D transduction, using adenoviral vectors, is superior to standard transduction protocols in 2D. It therefore, might help increasing its administration in tissue engineering and clinical applications.


Assuntos
Hidrogéis/química , Células-Tronco Mesenquimais/metabolismo , Alicerces Teciduais/química , Transdução Genética/métodos , Transfecção/métodos , Idoso , Alginatos/química , Proteína Morfogenética Óssea 2/análise , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Técnicas de Cultura de Células/métodos , Células Cultivadas , Feminino , Fibrina/química , Terapia Genética , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Humanos , Fator de Crescimento Insulin-Like I/análise , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade , Sefarose/química , Engenharia Tecidual/métodos
13.
Biomaterials ; 34(5): 1478-87, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23182922

RESUMO

Articular cartilage maturation is the postnatal development process that adapts joint surfaces to their site-specific biomechanical demands. Maturation involves gross morphological changes that occur through a process of synchronised growth and resorption of cartilage and generally ends at sexual maturity. The inability to induce maturation in biomaterial constructs designed for cartilage repair has been cited as a major cause for their failure in producing persistent cell-based repair of joint lesions. The combination of growth factors FGF2 and TGFß1 induces accelerated articular cartilage maturation in vitro such that many molecular and morphological characteristics of tissue maturation are observable. We hypothesised that experimental growth factor-induced maturation of immature cartilage would result in a biophysical and biochemical composition consistent with a mature phenotype. Using native immature and mature cartilage as reference, we observed that growth factor-treated immature cartilages displayed increased nano-compressive stiffness, decreased surface adhesion, decreased water content, increased collagen content and smoother surfaces, correlating with a convergence to the mature cartilage phenotype. Furthermore, increased gene expression of surface structural protein collagen type I in growth factor-treated explants compared to reference cartilages demonstrates that they are still in the dynamic phase of the postnatal developmental transition. These data provide a basis for understanding the regulation of postnatal maturation of articular cartilage and the application of growth factor-induced maturation in vitro and in vivo in order to repair and regenerate cartilage defects.


Assuntos
Cartilagem Articular/crescimento & desenvolvimento , Fator 2 de Crescimento de Fibroblastos/farmacologia , Técnicas de Cultura de Órgãos/métodos , Engenharia Tecidual/métodos , Fator de Crescimento Transformador beta2/farmacologia , Animais , Cartilagem Articular/efeitos dos fármacos , Bovinos , Masculino
14.
Eur Spine J ; 21(7): 1396-409, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22407261

RESUMO

INTRODUCTION: The basis of disc degeneration is still unknown, but is believed to be a cell-mediated process. Apoptosis might play a major role in degenerative disc disease (DDD). The aim of this study was to correlate the viability of disc cells with the radiological degeneration grades (rDG) in disc herniation. MATERIALS AND METHODS: Forty anterior IVD's (C4-C7) from 39 patients with DDD were studied histologically and ultrastructurally to quantify healthy, "balloon", chondroptotic, apoptotic and necrotic cells. Patients were classified to their rDG, as having either prolapse (P: DGII + III) and/or osteochondrosis (O: DGIV + V). Similar studies were undertaken on eight control discs. RESULTS: Cell death by necrosis (mean 35%) was common but differed not significantly in both groups. All patients with a disc prolapse DGII + III revealed balloon cells (iAF: mean 32%). All appeared alive and sometimes were hypertrophic. However, significantly less balloon cells were found in the O-Group. Control samples revealed no evidence of "balloon" cells in DGII and only a minor rate in DGIII. CONCLUSION: According to the different rDG, quantitative changes were obvious in healthy and "balloon" cells, but not for cell death. At the moment it can only be hypothesized if "balloon" cells are part of a repair strategy and/or cause of disc herniation.


Assuntos
Vértebras Cervicais/patologia , Vértebras Cervicais/ultraestrutura , Degeneração do Disco Intervertebral/patologia , Deslocamento do Disco Intervertebral/patologia , Disco Intervertebral/patologia , Disco Intervertebral/ultraestrutura , Adulto , Idoso , Apoptose , Cadáver , Estudos de Casos e Controles , Sobrevivência Celular , Vértebras Cervicais/diagnóstico por imagem , Feminino , Humanos , Disco Intervertebral/diagnóstico por imagem , Degeneração do Disco Intervertebral/diagnóstico por imagem , Deslocamento do Disco Intervertebral/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Necrose , Tomografia Computadorizada por Raios X
15.
Vet J ; 192(3): 345-51, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21968294

RESUMO

A chondrocyte progenitor population isolated from the surface zone of articular cartilage presents a promising cell source for cell-based cartilage repair. In this study, equine articular cartilage progenitor cells (ACPCs) and equine bone marrow-derived stromal cells (BMSCs) were compared as potential cell sources for repair. Clonally derived BMSCs and ACPCs demonstrated expression of the cell fate selector gene, Notch-1, and the putative stem cell markers STRO-1, CD90 and CD166. Chondrogenic induction revealed positive labelling for collagen type II and aggrecan. Collagen type X was not detected in ACPC pellets but was observed in all BMSC pellets. In addition, it was observed that BMSCs labelled for Runx2 and matrilin-1 antibodies, whereas ACPC labelling was significantly less or absent. For both cell types, osteogenic induction revealed positive von Kossa staining in addition to positive labelling for osteocalcin. Adipogenic induction revealed a positive result via oil red O staining in both cell types. ACPCs and BMSCs have demonstrated functional equivalence in their multipotent differentiation capacity. Chondrogenic induction of BMSCs resulted in a hypertrophic cartilage (endochondral) phenotype, which can limit cartilage repair as the tissue can undergo mineralisation. ACPCs may therefore be considered superior to BMSCs in producing cartilage capable of functional repair.


Assuntos
Células da Medula Óssea/citologia , Cartilagem Articular/citologia , Terapia Baseada em Transplante de Células e Tecidos/veterinária , Cavalos/fisiologia , Células-Tronco/citologia , Células Estromais/citologia , Animais , Células da Medula Óssea/fisiologia , Cartilagem Articular/fisiologia , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Colágeno/classificação , Colágeno/metabolismo , Regulação da Expressão Gênica/fisiologia , Imuno-Histoquímica/veterinária , Células-Tronco/fisiologia , Células Estromais/fisiologia
16.
Arthritis Rheum ; 63(11): 3417-27, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21769844

RESUMO

OBJECTIVE: We have discovered that a combination of fibroblast growth factor 2 and transforming growth factor ß1 induce profound morphologic changes in immature articular cartilage. The purpose of this study was to test the hypothesis that these changes represent accelerated postnatal maturation. METHODS: Histochemical and biochemical assays were used to confirm the nature of the morphologic changes that accompany growth factor stimulation of immature bovine articular cartilage explants in serum-free culture medium. Growth factor-induced apoptosis, cellular proliferation, and changes in the collagen network were also quantitatively analyzed. RESULTS: Growth factor stimulation resulted in rapid resorption from the basal aspect of immature cartilage explants that was simultaneously opposed by cellular proliferation from the apical aspect driven from a pool of chondroprogenitor cells we have previously described. Maturation-dependent changes in tissue stiffness, collagen crosslinking, and collagen fibril architecture as well as differentiation of the extracellular matrix into distinct pericellular, territorial, and interterritorial domains were all present in growth factor-stimulated cartilage samples and absent in control samples. CONCLUSION: Our data demonstrate that it is possible to significantly enhance the maturation of cartilage tissue using specific growth factor stimulation. This may have applications in transplantation therapy or in the treatment of diseased cartilage, through phenotype modulation of osteoarthritic chondrocytes in order to stimulate growth and maturation of cartilage repair tissue.


Assuntos
Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/crescimento & desenvolvimento , Condrócitos/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Apoptose/efeitos dos fármacos , Cartilagem Articular/metabolismo , Bovinos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Condrócitos/metabolismo , Colágeno/metabolismo , Masculino
17.
PLoS One ; 5(10): e13246, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-20976230

RESUMO

BACKGROUND: Articular cartilage displays a poor repair capacity. The aim of cell-based therapies for cartilage defects is to repair damaged joint surfaces with a functional replacement tissue. Currently, chondrocytes removed from a healthy region of the cartilage are used but they are unable to retain their phenotype in expanded culture. The resulting repair tissue is fibrocartilaginous rather than hyaline, potentially compromising long-term repair. Mesenchymal stem cells, particularly bone marrow stromal cells (BMSC), are of interest for cartilage repair due to their inherent replicative potential. However, chondrocyte differentiated BMSCs display an endochondral phenotype, that is, can terminally differentiate and form a calcified matrix, leading to failure in long-term defect repair. Here, we investigate the isolation and characterisation of a human cartilage progenitor population that is resident within permanent adult articular cartilage. METHODS AND FINDINGS: Human articular cartilage samples were digested and clonal populations isolated using a differential adhesion assay to fibronectin. Clonal cell lines were expanded in growth media to high population doublings and karyotype analysis performed. We present data to show that this cell population demonstrates a restricted differential potential during chondrogenic induction in a 3D pellet culture system. Furthermore, evidence of high telomerase activity and maintenance of telomere length, characteristic of a mesenchymal stem cell population, were observed in this clonal cell population. Lastly, as proof of principle, we carried out a pilot repair study in a goat in vivo model demonstrating the ability of goat cartilage progenitors to form a cartilage-like repair tissue in a chondral defect. CONCLUSIONS: In conclusion, we propose that we have identified and characterised a novel cartilage progenitor population resident in human articular cartilage which will greatly benefit future cell-based cartilage repair therapies due to its ability to maintain chondrogenicity upon extensive expansion unlike full-depth chondrocytes that lose this ability at only seven population doublings.


Assuntos
Cartilagem Articular/citologia , Células-Tronco/citologia , Adolescente , Adulto , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Criança , Primers do DNA , Fibronectinas/química , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Telomerase/metabolismo , Telômero
18.
Stem Cells Dev ; 19(6): 819-30, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20132052

RESUMO

Wounds within the oral mucosa, similarly to fetal wounds, exhibit rapid healing with reduced scarring. We hypothesized that a progenitor population resident within the oral mucosal lamina propria (OMLP) contributes to this preferential healing. Progenitor cells (PC) were reliably isolated from the OMLP by differential adhesion to fibronectin. Isolated colonies originating from a single cell demonstrated a rapid initial phase of proliferation, completing in excess of 50 population doublings (PDs) before entering cellular senescence. These data were supported by the expression of active telomerase within both developing colonies and expanded clones as assessed by immunocytochemistry (ICC) and the quantitative telomeric repeat amplification protocol. FACS analysis confirmed expression of the stem cell markers CD44, CD90, CD105, and CD166, but negative expression of CD34 and CD45 ruling out a hematopoietic or fibrocyte origin for these progenitors. A neural crest origin was confirmed by increased colony-forming efficiency (CFE) in the presence of Jagged 1 and the expression of a number of neural crest markers within the developing colonies by ICC and serially passaged clones by Western blotting. The multipotency of this novel PC population was demonstrated by differentiation of the cells down both mesenchymal (chondrogenic, osteoblastic, and adipogenic) and neuronal (neuron and Schwann-like cells) cell lineages. This article reports for the first time, the isolation and characterization of a novel, clonally derived PC population resident within the OMLP. The attributes of this adult stem cell (ASC) population and its accessibility lends itself to future therapeutic applications.


Assuntos
Mucosa Bucal/citologia , Células-Tronco Multipotentes/citologia , Crista Neural/citologia , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Membrana Celular/metabolismo , Proliferação de Células , Separação Celular , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mucosa Bucal/enzimologia , Células-Tronco Multipotentes/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Telomerase/metabolismo
19.
Eur Cell Mater ; 19: 1-12, 2010 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-20077400

RESUMO

Chick wing bud mesenchymal cell micromass culture allows the study of a variety of developmental mechanisms, ranging from cell adhesion to pattern formation. However, many cells remain in contact with an artificial substratum, which can influence cytoskeletal organisation and differentiation. An ultrasound standing wave trap facilitates the rapid formation of 2-D monolayer cell aggregates with a defined zero time-point, independent from contact with a surface. Aggregates formed rapidly (within 2 min) and intercellular membrane spreading occurred at points of contact. This was associated with an increase in peripheral F-actin within 10 min of cell-cell contact and aggregates had obtained physical integrity by 30 min. The chondrogenic transcription factor Sox9 could be detected in cells in the ultrasound trap within 3 h (ultrasound exposure alone was not responsible for this effect). This approach facilitates the study of the initial cell-cell interactions that occur during condensation formation and demonstrates that a combination of cell shape and cytoskeletal organisation is required for the initiation and maintenance of a differentiated phenotype, which is lost when these phenomena are influenced by contact with an artificial substrate.


Assuntos
Condrogênese , Células-Tronco Mesenquimais/citologia , Modelos Biológicos , Ultrassom , Asas de Animais/citologia , Asas de Animais/embriologia , Actinas/metabolismo , Animais , Agregação Celular , Núcleo Celular/metabolismo , Forma Celular , Embrião de Galinha , Crioultramicrotomia , Citoesqueleto/metabolismo , Fibroblastos/citologia , Transporte Proteico , Fatores de Transcrição SOX9/metabolismo
20.
Microsc Res Tech ; 73(8): 741-5, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19953665

RESUMO

Immunohistochemical detection of cross-linked fibrillar collagens in mineralized tissues is much desired for exploring the mechanisms of biomineralization in health and disease. Mineralized teeth are impossible to section when embedded in conventional media, thus limiting on-section characterization of matrix proteins by immunohistochemistry. We hypothesized that by using an especially formulated acrylic resin suitable for mineralized dental tissues, not only sectioning of teeth would be possible, but also our recently developed immunofluorescence labeling technique would be amenable to fully calcified tissues for characterization of dentinal fibrillar collagens, which remains elusive. The hypothesis was tested on fixed rodent teeth embedded in Technovit 9100 New. It was possible to cut thin (1 mum) sections of mineralized teeth, and immunofluorescence characterization of cross-linked type I fibrillar collagen was selected due to its abundance in dentine. Decalcified samples of teeth embedded in paraffin wax were also used to compare immunolabeling from either method using the same immunoreagents in equivalent concentrations. In the decalcified tissue sections, type I collagen labeling in the dentine along the tubules was "patchy" and the signal in the predentine was very weak. However, enhanced signal in mineralized samples with type I collagen was detected not only in the predentine but also at the limit between intertubular dentine, within the elements of the enamel organ and subgingival stroma. This report offers advances in sectioning mineralized dental tissues and allows the application of immunofluorescence not only for on-section protein detection but importantly for detecting cross-linked fibrous collagens in both soft and mineralized tissue sections.


Assuntos
Técnicas de Preparação Histocitológica/métodos , Calcificação de Dente , Dente/ultraestrutura , Resinas Acrílicas , Animais , Colágeno Tipo I/análise , Dentina/química , Dentina/ultraestrutura , Fixadores , Imunofluorescência , Imuno-Histoquímica , Masculino , Microscopia , Inclusão em Parafina , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...