Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nat Rev Rheumatol ; 19(7): 403-416, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37296196

RESUMO

Osteoarthritis (OA) is a disabling condition that affects billions of people worldwide and places a considerable burden on patients and on society owing to its prevalence and economic cost. As cartilage injuries are generally associated with the progressive onset of OA, robustly effective approaches for cartilage regeneration are necessary. Despite extensive research, technical development and clinical experimentation, no current surgery-based, material-based, cell-based or drug-based treatment can reliably restore the structure and function of hyaline cartilage. This paucity of effective treatment is partly caused by a lack of fundamental understanding of why articular cartilage fails to spontaneously regenerate. Thus, research studies that investigate the mechanisms behind the cartilage regeneration processes and the failure of these processes are critical to instruct decisions about patient treatment or to support the development of next-generation therapies for cartilage repair and OA prevention. This Review provides a synoptic and structured analysis of the current hypotheses about failure in cartilage regeneration, and the accompanying therapeutic strategies to overcome these hurdles, including some current or potential approaches to OA therapy.


Assuntos
Cartilagem Articular , Osteoartrite , Humanos , Condrócitos , Osteoartrite/terapia , Regeneração
2.
Stem Cells ; 38(11): 1438-1453, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32652878

RESUMO

Mesenchymal stem cells (MSCs) have been investigated as a potential injectable therapy for the treatment of knee osteoarthritis, with some evidence of success in preliminary human trials. However, optimization and scale-up of this therapeutic approach depends on the identification of functional markers that are linked to their mechanism of action. One possible mechanism is through their chondrogenic differentiation and direct role in neo-cartilage synthesis. Alternatively, they could remain undifferentiated and act through the release of trophic factors that stimulate endogenous repair processes within the joint. Here, we show that extensive in vitro aging of bone marrow-derived human MSCs leads to loss of chondrogenesis but no reduction in trophic repair, thereby separating out the two modes of action. By integrating transcriptomic and proteomic data using Ingenuity Pathway Analysis, we found that reduced chondrogenesis with passage is linked to downregulation of the FOXM1 signaling pathway while maintenance of trophic repair is linked to CXCL12. In an attempt at developing functional markers of MSC potency, we identified loss of mRNA expression for MMP13 as correlating with loss of chondrogenic potential of MSCs and continued secretion of high levels of TIMP1 protein as correlating with the maintenance of trophic repair capacity. Since an allogeneic injectable osteoar therapy would require extensive cell expansion in vitro, we conclude that early passage MMP13+ , TIMP1-secretinghigh MSCs should be used for autologous OA therapies designed to act through engraftment and chondrogenesis, while later passage MMP13- , TIMP1-secretinghigh MSCs could be exploited for allogeneic OA therapies designed to act through trophic repair.


Assuntos
Metaloproteinase 13 da Matriz/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Osteoartrite/terapia , Engenharia Tecidual/métodos , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo
3.
Stem Cells ; 35(11): 2280-2291, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28833807

RESUMO

Multipotent mesenchymal stem cells (MSCs) have enormous potential in tissue engineering and regenerative medicine. However, until now, their development for clinical use has been severely limited as they are a mixed population of cells with varying capacities for lineage differentiation and tissue formation. Here, we identify receptor tyrosine kinase-like orphan receptor 2 (ROR2) as a cell surface marker expressed by those MSCs with an enhanced capacity for cartilage formation. We generated clonal human MSC populations with varying capacities for chondrogenesis. ROR2 was identified through screening for upregulated genes in the most chondrogenic clones. When isolated from uncloned populations, ROR2+ve MSCs were significantly more chondrogenic than either ROR2-ve or unfractionated MSCs. In a sheep cartilage-repair model, they produced significantly more defect filling with no loss of cartilage quality compared with controls. ROR2+ve MSCs/perivascular cells were present in developing human cartilage, adult bone marrow, and adipose tissue. Their frequency in bone marrow was significantly lower in patients with osteoarthritis (OA) than in controls. However, after isolation of these cells and their initial expansion in vitro, there was greater ROR2 expression in the population derived from OA patients compared with controls. Furthermore, osteoarthritis-derived MSCs were better able to form cartilage than MSCs from control patients in a tissue engineering assay. We conclude that MSCs expressing high levels of ROR2 provide a defined population capable of predictably enhanced cartilage production. Stem Cells 2017;35:2280-2291.


Assuntos
Condrogênese/genética , Células-Tronco Mesenquimais/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Proteína Wnt-5a/genética , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Humanos , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Ovinos , Engenharia Tecidual , Proteína Wnt-5a/metabolismo
4.
Stem Cells Transl Med ; 6(4): 1237-1248, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28186682

RESUMO

Meniscal cartilage tears are common and predispose to osteoarthritis (OA). Most occur in the avascular portion of the meniscus where current repair techniques usually fail. We described previously the use of undifferentiated autologous mesenchymal stem cells (MSCs) seeded onto a collagen scaffold (MSC/collagen-scaffold) to integrate meniscal tissues in vitro. Our objective was to translate this method into a cell therapy for patients with torn meniscus, with the long-term goal of delaying or preventing the onset of OA. After in vitro optimization, we tested an ovine-MSC/collagen-scaffold in a sheep meniscal cartilage tear model with promising results after 13 weeks, although repair was not sustained over 6 months. We then conducted a single center, prospective, open-label first-in-human safety study of patients with an avascular meniscal tear. Autologous MSCs were isolated from an iliac crest bone marrow biopsy, expanded and seeded into the collagen scaffold. The resulting human-MSC/collagen-scaffold implant was placed into the meniscal tear prior to repair with vertical mattress sutures and the patients were followed for 2 years. Five patients were treated and there was significant clinical improvement on repeated measures analysis. Three were asymptomatic at 24 months with no magnetic resonance imaging evidence of recurrent tear and clinical improvement in knee function scores. Two required subsequent meniscectomy due to retear or nonhealing of the meniscal tear at approximately 15 months after implantation. No other adverse events occurred. We conclude that undifferentiated MSCs could provide a safe way to augment avascular meniscal repair in some patients. Registration: EU Clinical Trials Register, 2010-024162-22. Stem Cells Translational Medicine 2017;6:1237-1248.


Assuntos
Doenças das Cartilagens/terapia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Lesões do Menisco Tibial/terapia , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Feminino , Humanos , Técnicas In Vitro , Meniscos Tibiais/citologia , Ovinos , Engenharia Tecidual/métodos , Alicerces Teciduais , Cicatrização/fisiologia
5.
Nat Commun ; 6: 7405, 2015 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-26080734

RESUMO

Restricted oxygen diffusion can result in central cell necrosis in engineered tissue, a problem that is exacerbated when engineering large tissue constructs for clinical application. Here we show that pre-treating human mesenchymal stem cells (hMSCs) with synthetic membrane-active myoglobin-polymer-surfactant complexes can provide a reservoir of oxygen capable of alleviating necrosis at the centre of hyaline cartilage. This is achieved through the development of a new cell functionalization methodology based on polymer-surfactant conjugation, which allows the delivery of functional proteins to the hMSC membrane. This new approach circumvents the need for cell surface engineering using protein chimerization or genetic transfection, and we demonstrate that the surface-modified hMSCs retain their ability to proliferate and to undergo multilineage differentiation. The functionalization technology is facile, versatile and non-disruptive, and in addition to tissue oxygenation, it should have far-reaching application in a host of tissue engineering and cell-based therapies.


Assuntos
Cartilagem Hialina , Células-Tronco Mesenquimais/efeitos dos fármacos , Mioglobina/farmacologia , Oxigênio/administração & dosagem , Engenharia Tecidual/métodos , Escherichia coli , Glicolatos/química , Humanos , Mioglobina/química
6.
Cartilage ; 6(2 Suppl): 30S-5S, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27340514

RESUMO

Chondrogenic progenitor populations, including mesenchymal stem cells, represent promising cell-based transplantation or tissue engineering therapies for the regeneration of damaged cartilage. Osteoarthritis (OA) predominantly affects the elderly and is a leading cause of disability worldwide. Advancing age is a prominent risk factor that is closely associated with the onset and progression of the disease. Understanding the influence that aging and OA have on chondrogenic progenitor cells is important to determine how these processes affect the cellular mechanisms of the cells and their capacity to differentiate into functional chondrocytes for use in therapeutic applications. Here, we review the effect of age- and OA-related changes on the growth kinetics and differentiation potential of chondrogenic progenitor cell populations. Aging differentially influences the proliferative potential of progenitor cells showing reduced growth rates with increased senescence and apoptotic activity over time, while chondrogenesis appears to be independent of donor age. Cartilage tissue affected by OA shows evidence of progenitor populations with some potential for repair, however reports on the proliferative propensity of mesenchymal stem cells and their chondrogenic potential are contradictory. This is likely attributed to the narrow age ranges of samples assessed and deficits in definitively identifying donors with OA versus healthy patients across a wide scope of advancing ages. Further studies that investigate the mechanistic effects of chondrogenic progenitor populations associated with aging and the progression of OA using clearly defined criteria and age-matched control subject groups are crucial to our understanding of the clinical relevance of these cells for use in cartilage repair therapies.

7.
Stem Cells Dev ; 23(5): 541-54, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24172175

RESUMO

Cartilage injuries and osteoarthritis are leading causes of disability in developed countries. The regeneration of damaged articular cartilage using cell transplantation or tissue engineering holds much promise but requires the identification of an appropriate cell source with a high proliferative propensity and consistent chondrogenic capacity. Human fetal mesenchymal stem cells (MSCs) have been isolated from a range of perinatal tissues, including first-trimester bone marrow, and have demonstrated enhanced expansion and differentiation potential. However, their ability to form mature chondrocytes for use in cartilage tissue engineering has not been clearly established. Here, we compare the chondrogenic potential of human MSCs isolated from fetal and adult bone marrow and show distinct differences in their responsiveness to specific growth factors. Transforming growth factor beta 3 (TGFß3) induced chondrogenesis in adult but not fetal MSCs. In contrast, bone morphogenetic protein 2 (BMP2) induced chondrogenesis in fetal but not adult MSCs. When fetal MSCs co-stimulated with BMP2 and TGFß3 were used for cartilage tissue engineering, they generated tissue with type II collagen and proteoglycan content comparable to adult MSCs treated with TGFß3 alone. Investigation of the TGFß/BMP signaling pathway showed that TGFß3 induced phosphorylation of SMAD3 in adult but not fetal MSCs. These findings demonstrate that the initiation of chondrogenesis is modulated by distinct signaling mechanisms in fetal and adult MSCs. This study establishes the feasibility of using fetal MSCs in cartilage repair applications and proposes their potential as an in vitro system for modeling chondrogenic differentiation and skeletal development studies.


Assuntos
Diferenciação Celular/genética , Condrogênese/genética , Células-Tronco Mesenquimais/citologia , Transdução de Sinais/genética , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Células da Medula Óssea/citologia , Cartilagem/patologia , Cartilagem/transplante , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Engenharia Tecidual , Fator de Crescimento Transformador beta3
8.
Biomacromolecules ; 14(5): 1287-98, 2013 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-23534615

RESUMO

Biomaterials that can stimulate stem cell differentiation without growth factor supplementation provide potent and cost-effective scaffolds for regenerative medicine. We hypothesize that a scaffold prepared from cellulose and silk blends can direct stem cell chondrogenic fate. We systematically prepared cellulose blends with silk at different compositions using an environmentally benign processing method based on ionic liquids as a common solvent. We tested the effect of blend compositions on the physical properties of the materials as well as on their ability to support mesenchymal stem cell (MSC) growth and chondrogenic differentiation. The stiffness and tensile strength of cellulose was significantly reduced by blending with silk. The characterized materials were tested using MSCs derived from four different patients. Growing MSCs on a specific blend combination of cellulose and silk in a 75:25 ratio significantly upregulated the chondrogenic marker genes SOX9, aggrecan, and type II collagen in the absence of specific growth factors. This chondrogenic effect was neither found with neat cellulose nor the cellulose/silk 50:50 blend composition. No adipogenic or osteogenic differentiation was detected on the blends, suggesting that the cellulose/silk 75:25 blend induced specific stem cell differentiation into the chondrogenic lineage without addition of the soluble growth factor TGF-ß. The cellulose/silk blend we identified can be used both for in vitro tissue engineering and as an implantable device for stimulating endogenous stem cells to initiate cartilage repair.


Assuntos
Materiais Biocompatíveis/farmacologia , Celulose/química , Condrócitos/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Seda/química , Engenharia Tecidual/métodos , Agrecanas/genética , Agrecanas/metabolismo , Materiais Biocompatíveis/química , Diferenciação Celular , Condrócitos/citologia , Condrócitos/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Líquidos Iônicos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Resistência à Tração , Alicerces Teciduais
9.
Cartilage ; 2(2): 153-72, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26069577

RESUMO

Cartilage repair strategies aim to resurface a lesion with osteochondral tissue resembling native cartilage, but a variety of repair tissues are usually observed. Histology is an important structural outcome that could serve as an interim measure of efficacy in randomized controlled clinical studies. The purpose of this article is to propose guidelines for standardized histoprocessing and unbiased evaluation of animal tissues and human biopsies. Methods were compiled from a literature review, and illustrative data were added. In animal models, treatments are usually administered to acute defects created in healthy tissues, and the entire joint can be analyzed at multiple postoperative time points. In human clinical therapy, treatments are applied to developed lesions, and biopsies are obtained, usually from a subset of patients, at a specific time point. In striving to standardize evaluation of structural endpoints in cartilage repair studies, 5 variables should be controlled: 1) location of biopsy/sample section, 2) timing of biopsy/sample recovery, 3) histoprocessing, 4) staining, and 5) blinded evaluation with a proper control group. Histological scores, quantitative histomorphometry of repair tissue thickness, percentage of tissue staining for collagens and glycosaminoglycan, polarized light microscopy for collagen fibril organization, and subchondral bone integration/structure are all relevant outcome measures that can be collected and used to assess the efficacy of novel therapeutics. Standardized histology methods could improve statistical analyses, help interpret and validate noninvasive imaging outcomes, and permit cross-comparison between studies. Currently, there are no suitable substitutes for histology in evaluating repair tissue quality and cartilaginous character.

10.
Stem Cells ; 28(11): 1992-6, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20882533

RESUMO

Cartilage is considered to be a simple tissue that should be easy to engineer because it is avascular and contains just one cell type, the chondrocyte. Despite this apparent simplicity, regenerating cartilage in a form that can function effectively after implantation in the joint has proven difficult. This may be because we have not fully appreciated the importance of different structural regions of articular cartilage or of understanding the origins of chondrocytes and how this cell population is maintained in the normal tissue. This review considers what is known about different regions of cartilage and the types of stem cells in articulating joints and emphasizes the potential importance of regeneration of the lamina splendens at the joint surface and calcified cartilage at the junction with bone for long-term survival of regenerated tissue in vivo.


Assuntos
Cartilagem/citologia , Regeneração/fisiologia , Células-Tronco Adultas/citologia , Animais , Condrogênese/fisiologia , Humanos , Células-Tronco Mesenquimais/citologia , Modelos Biológicos
11.
Biomaterials ; 31(9): 2583-91, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20053438

RESUMO

Injuries to the avascular region of knee meniscal cartilage do not heal spontaneously. To address this problem we have developed a new stem cell/collagen-scaffold implant system in which human adult bone marrow mesenchymal stem cells are seeded onto a biodegradable scaffold that allows controlled delivery of actively dividing cells to the meniscus surface. Sandwich constructs of two white zone ovine meniscus discs with stem cell/collagen-scaffold implant in between were cultured in vitro for 40 days. Histomorphometric analysis revealed superior integration in the stem cell/collagen-scaffold groups compared to the cell-free collagen membrane or untreated controls. The addition of TGF-beta1 to differentiate stem cells to chondrocytes inhibited integration. Biomechanical testing demonstrated a significant 2-fold increase in tensile strength in all constructs using the stem cell/collagen-scaffold compared to control groups after 40 days in culture. Integration was significantly higher when collagen membranes were used that had a more open/spongy structure adjacent to both meniscal cartilage surfaces, whereas a collagen scaffold designed for osteoinduction failed to induce any integration of meniscus. In conclusion, the stem cell/collagen-scaffold implant is a potential therapeutic treatment for the repair of white zone meniscal cartilage tears.


Assuntos
Cartilagem/patologia , Colágeno/farmacologia , Implantes Experimentais , Meniscos Tibiais/patologia , Células-Tronco Mesenquimais/citologia , Alicerces Teciduais/química , Cicatrização/efeitos dos fármacos , Animais , Cartilagem/efeitos dos fármacos , Bovinos , Diferenciação Celular/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Humanos , Fenômenos Mecânicos/efeitos dos fármacos , Membranas Artificiais , Meniscos Tibiais/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Ovinos
12.
Biomaterials ; 30(29): 5260-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19647867

RESUMO

Cell and tissue engineering are now being translated into clinical organ replacement, offering alternatives to fight morbidity, organ shortages and ethico-social problems associated with allotransplantation. Central to the recent first successful use of stem cells to create an organ replacement in man was our development of a bioreactor environment. Critical design features were the abilities to drive the growth of two different cell types, to support 3D maturation, to maintain biomechanical and biological properties and to provide appropriate hydrodynamic stimuli and adequate mass transport. An analytical model was developed and applied to predict oxygen profiles in the bioreactor-cultured organ construct and in the culture media, comparing representative culture configurations and operating conditions. Autologous respiratory epithelial cells and mesenchymal stem cells (BMSCs, then differentiated into chondrocytes) were isolated, characterized and expanded. Both cell types were seeded and cultured onto a decellularized human donor tracheal matrix within the bioreactor. One year post-operatively, graft and patient are healthy, and biopsies confirm angiogenesis, viable epithelial cells and chondrocytes. Our rotating double-chamber bioreactor permits the efficient repopulation of a decellularized human matrix, a concept that can be applied clinically, as demonstrated by the successful tracheal transplantation.


Assuntos
Órgãos Bioartificiais , Reatores Biológicos , Células Epiteliais/transplante , Transplante de Células-Tronco Mesenquimais/métodos , Técnicas de Cultura de Órgãos/instrumentação , Engenharia Tecidual/instrumentação , Traqueia/crescimento & desenvolvimento , Traqueia/transplante , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Desenho de Equipamento , Análise de Falha de Equipamento , Humanos , Rotação , Resultado do Tratamento
13.
Arthritis Res Ther ; 10(6): R132, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19014452

RESUMO

INTRODUCTION: The present study established characteristics of tissue regrowth in patients suffering knee lesions treated with grafts of autologous chondrocytes grown on three-dimensional hyaluronic acid biomaterials. METHODS: This multicentred study involved a second-look arthroscopy/biopsy, 5 to 33 months post implant (n = 63). Seven patients allowed a third-look biopsy, three of which were performed 18 months post implant. Characteristics of tissues were histologically and histochemically evaluated. The remaining bone stubs were evaluated for cartilage/bone integration. For data analysis, biopsies were further divided into those obtained from postoperative symptomatic patients (n = 41) or from asymptomatic patients (n = 22). RESULTS: The percentage of hyaline regenerated tissues was significantly greater in biopsies obtained after, versus within, 18 months of implantation. Differences were also observed between symptomatic and asymptomatic patients: reparative tissues taken from symptomatic patients 18 months after grafting were mainly fibrocartilage or mixed (hyaline-fibrocartilage) tissue, while tissues taken from asymptomatic patients were hyaline cartilage in 83% of biopsies. In a small group of asymptomatic patients (n = 3), second-look and third-look biopsies taken 18 months after surgery confirmed maturation of the newly formed tissue over time. Cartilage maturation occurred from the inner regions of the graft, in contact with subchondral bone, towards the periphery of the implant. CONCLUSIONS: The study indicates that, in asymptomatic patients after chondrocyte implantation, regenerated tissue undergoes a process of maturation that in the majority of cases takes longer than 18 months for completion and leads to hyaline tissue and not fibrous cartilage. Persistence of symptoms might reflect the presence of a nonhyaline cartilage repair tissue.


Assuntos
Cartilagem Articular/transplante , Cirurgia de Second-Look/métodos , Engenharia Tecidual/métodos , Cicatrização/fisiologia , Adolescente , Adulto , Biópsia , Cartilagem Articular/química , Estudos de Coortes , Feminino , Seguimentos , Humanos , Articulação do Joelho/química , Articulação do Joelho/patologia , Articulação do Joelho/cirurgia , Masculino , Pessoa de Meia-Idade , Fatores de Tempo , Transplante de Tecidos/métodos , Adulto Jovem
14.
Lancet ; 372(9655): 2023-30, 2008 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-19022496

RESUMO

BACKGROUND: The loss of a normal airway is devastating. Attempts to replace large airways have met with serious problems. Prerequisites for a tissue-engineered replacement are a suitable matrix, cells, ideal mechanical properties, and the absence of antigenicity. We aimed to bioengineer tubular tracheal matrices, using a tissue-engineering protocol, and to assess the application of this technology in a patient with end-stage airway disease. METHODS: We removed cells and MHC antigens from a human donor trachea, which was then readily colonised by epithelial cells and mesenchymal stem-cell-derived chondrocytes that had been cultured from cells taken from the recipient (a 30-year old woman with end-stage bronchomalacia). This graft was then used to replace the recipient's left main bronchus. FINDINGS: The graft immediately provided the recipient with a functional airway, improved her quality of life, and had a normal appearance and mechanical properties at 4 months. The patient had no anti-donor antibodies and was not on immunosuppressive drugs. INTERPRETATION: The results show that we can produce a cellular, tissue-engineered airway with mechanical properties that allow normal functioning, and which is free from the risks of rejection. The findings suggest that autologous cells combined with appropriate biomaterials might provide successful treatment for patients with serious clinical disorders.


Assuntos
Broncomalácia/fisiopatologia , Condrócitos/citologia , Engenharia Tecidual/métodos , Traqueia/transplante , Adulto , Broncomalácia/terapia , Cadáver , Feminino , Humanos , Período Pós-Operatório , Testes de Função Respiratória , Traqueia/citologia
15.
Stem Cells ; 25(10): 2460-8, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17615267

RESUMO

Conventional methods for regulating the differentiation of stem cells are largely based on the use of biological agents such as growth factors. We hypothesize that stem cell differentiation could be driven by specific synthetic molecules. If true, this would offer the possibility of screening chemical libraries to develop pharmacological agents with improved efficacy. To test our hypothesis, we have determined which, if any, of the nuclear receptor superfamily might be involved in chondrogenesis. We used fluorescence-activated cell sorting, as well as quantitative polymerase chain reaction, to study expression of a range of nuclear receptors in the undifferentiated mesenchymal population and after growth factor-driven differentiation of these cells to chondrocytes. In this way, we identified retinoic acid receptor beta (RAR beta) as a potential pharmacological target. A low molecular weight synthetic inhibitor of the RAR alpha and RAR beta receptors was able to induce chondrogenic differentiation of mesenchymal stem cells derived from osteoarthritis patients, in the absence of serum and growth factors. Furthermore, the pathway is independent of SOX9 upregulation and does not lead to hypertrophy. When mesenchymal cells were seeded on to polyglycolic acid scaffolds and cultured with LE135, there was a dose-dependent formation of cartilage, demonstrated both histologically and by biochemical analysis of the collagen component of the extracellular matrix. These results demonstrate the feasibility of a pharmacological approach to the regulation of stem cell function. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Células-Tronco Adultas/efeitos dos fármacos , Condrócitos/citologia , Condrogênese/efeitos dos fármacos , Dibenzazepinas/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Receptores do Ácido Retinoico/antagonistas & inibidores , Adipócitos/citologia , Adulto , Células-Tronco Adultas/citologia , Idoso , Cartilagem Articular/citologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Condrogênese/genética , Meios de Cultura Livres de Soro/farmacologia , Feminino , Proteínas de Grupo de Alta Mobilidade/antagonistas & inibidores , Proteínas de Grupo de Alta Mobilidade/fisiologia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade , Osteoartrite/patologia , Osteócitos/citologia , RNA Interferente Pequeno/farmacologia , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Citoplasmáticos e Nucleares/genética , Receptores do Ácido Retinoico/fisiologia , Fatores de Transcrição SOX9 , Engenharia Tecidual/métodos , Alicerces Teciduais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/fisiologia
16.
Knee Surg Sports Traumatol Arthrosc ; 15(5): 610-9, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17372718

RESUMO

A biodegradable, hyaluronian-based biocompatible scaffold was used for autologous chondrocyte transplantation. This prospective study analyzes a clinical outcome of 70 consecutive patients treated by arthroscopic autologous chondrocyte transplantation at minimum 24 months follow up (47 of these patients achieved minimum 36 months follow-up and 21 patients minimum 48 months follow-up) in order to establish clear indication criteria for this type of treatment. 31 of these patients presented isolated chondral lesions, while 39 patients with associated lesions (23 ACL lesions, 28 meniscal lesions, 1 varus knee) were treated during the same surgical procedure with cartilage harvesting. A statistically significant clinical improvement was shown just at 24 months and the second-look arthroscopy demonstrated a complete coverage of the grafted area with a hyaline cartilage-like tissue in 12 of 15 analyzed patients. A better clinical outcome was observed in young, well-trained patients and in traumatic lesions. Other factors, such as defect size, localization, previous and associated surgery did not influence significantly the results. This matrix autologous chondrocyte transplantation procedure simplifies the surgical procedure and can be performed arthroscopically, thus reducing surgical morbidity and recovery time.


Assuntos
Cartilagem Articular/cirurgia , Condrócitos/transplante , Traumatismos do Joelho/cirurgia , Adjuvantes Imunológicos/farmacologia , Adolescente , Adulto , Artroscopia , Cartilagem Articular/lesões , Células Cultivadas/transplante , Condrócitos/citologia , Seguimentos , Humanos , Ácido Hialurônico/farmacologia , Traumatismos do Joelho/reabilitação , Pessoa de Meia-Idade , Seleção de Pacientes , Estudos Prospectivos , Transplante Autólogo
17.
Arthritis Rheum ; 56(1): 177-87, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17195220

RESUMO

OBJECTIVE: To determine whether it is possible to engineer 3-dimensional hyaline cartilage using mesenchymal stem cells derived from the bone marrow (BMSCs) of patients with osteoarthritis (OA). METHODS: Expanded BMSCs derived from patients with hip OA were seeded onto polyglycolic acid scaffolds and differentiated using transforming growth factor beta3 in the presence or absence of parathyroid hormone-related protein (PTHrP) to regulate hypertrophy. Micromass pellet cultures were established using the same cells for comparison. At the end of culture, the constructs or pellets were processed for messenger RNA (mRNA) analysis by quantitative real-time reverse transcription-polymerase chain reaction. Matrix proteins were analyzed using specific assays. RESULTS: Cartilage constructs engineered from BMSCs were at least 5 times the weight of equivalent pellet cultures. Histologic, mRNA, and biochemical analyses of the constructs showed extensive synthesis of proteoglycan and type II collagen but only low levels of type I collagen. The protein content was almost identical to that of cartilage engineered from bovine nasal chondrocytes. Analysis of type X collagen mRNA revealed a high level of mRNA in chondrogenic constructs compared with that in undifferentiated BMSCs, indicating an increased risk of hypertrophy in the tissue-engineered cells. However, the inclusion of PTHrP at a dose of 1 microM or 10 microM during the culture period resulted in significant suppression of type X collagen mRNA expression and a significant decrease in alkaline phosphatase activity, without any loss of the cartilage-specific matrix proteins. CONCLUSION: Three-dimensional hyaline cartilage can be engineered using BMSCs from patients with OA. This method could thus be used for the repair of cartilage lesions.


Assuntos
Cartilagem Hialina/citologia , Células-Tronco Mesenquimais/citologia , Osteoartrite do Quadril/patologia , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Engenharia Tecidual/métodos , Fatores de Crescimento Transformadores/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Bovinos , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Cartilagem Hialina/metabolismo , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Pessoa de Meia-Idade , Proteoglicanas/genética , Proteoglicanas/metabolismo , RNA Mensageiro/metabolismo
18.
Ann Surg ; 244(6): 978-85; discussion 985, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17122623

RESUMO

OBJECTIVE: To investigate if precultivation of human engineered nasal cartilage grafts of clinically relevant size would increase the suture retention strength at implantation and the tensile and bending stiffness 2 weeks after implantation. SUMMARY BACKGROUND INFORMATION: To be used for reconstruction of nasal cartilage defects, engineered grafts need to be reliably sutured at implantation and resist to bending/tension forces about 2 weeks after surgery, when fixation is typically removed. METHODS: Nasal septum chondrocytes from 4 donors were expanded for 2 passages and statically loaded on 15 x 5 x 2-mm size nonwoven meshes of esterified hyaluronan (Hyaff-11). Constructs were implanted for 2 weeks in nude mice between muscle fascia and subcutaneous tissue either directly after cell seeding or after 2 or 4 weeks of preculture in chondrogenic medium. Engineered tissues and native nasal cartilage were assessed histologically, biochemically, and biomechanically. RESULTS: Engineered constructs reproducibly developed with culture time into cartilaginous tissues with increasing content of glycosaminoglycans and collagen type II. Suture retention strength was significantly higher (3.6 +/- 2.2-fold) in 2-week precultured constructs than in freshly seeded meshes. Following in vivo implantation, tissues further developed and maintained the original scaffold size and shape. The bending stiffness was significantly higher (1.8 +/- 0.8-fold) if constructs were precultured for 2 weeks than if they were directly implanted, whereas tensile stiffness was close to native cartilage in all groups. CONCLUSION: In our experimental setup, preculture for 2 weeks was necessary to engineer nasal cartilage grafts with enhanced mechanical properties relevant for clinical use in facial reconstructive surgery.


Assuntos
Condrócitos/fisiologia , Septo Nasal/citologia , Rinoplastia , Técnicas de Cultura de Tecidos/métodos , Engenharia Tecidual , Adulto , Animais , Humanos , Ácido Hialurônico/análogos & derivados , Camundongos , Pessoa de Meia-Idade , Maleabilidade , Técnicas de Sutura , Resistência à Tração
19.
Tissue Eng ; 12(7): 1787-98, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16889509

RESUMO

The regeneration of damaged organs requires that engineered tissues mature when implanted at sites of injury or disease. We have used new analytic techniques to determine the extent of tissue regeneration after treatment of knee injury patients with a novel cartilage tissue engineering therapy and the effect of pre-existing osteoarthritis on the regeneration process. We treated 23 patients, with a mean age of 35.6 years, presenting with knee articular cartilage defects 1.5 cm2 to 11.25 cm2 (mean, 5.0 cm2) in area. Nine of the patients had X-ray evidence of osteoarthritis. Chondrocytes were isolated from healthy cartilage removed at arthroscopy. The cells were cultured for 14 days, seeded onto esterified hyaluronic acid scaffolds (Hyalograft C), and grown for a further 14 days before implantation. A second-look biopsy was taken from each patient after 6 to 30 months (mean, 16 months). After standard histological analysis, uncut tissue was further analyzed using a newly developed biochemical protocol involving digestion with trypsin and specific, quantitative assays for type II collagen, type I collagen, and proteoglycan, as well as mature and immature collagen crosslinks. Cartilage regeneration was observed as early as 11 months after implantation and in 10 out of 23 patients. Tissue regeneration was found even when implants were placed in joints that had already progressed to osteoarthrosis. Cartilage injuries can be effectively repaired using tissue engineering, and osteoarthritis does not inhibit the regeneration process.


Assuntos
Bioprótese , Cartilagem/transplante , Condrócitos/transplante , Ácido Hialurônico , Osteoartrite do Joelho/terapia , Regeneração , Engenharia Tecidual , Adolescente , Adulto , Cartilagem/metabolismo , Condrócitos/metabolismo , Proteínas da Matriz Extracelular/biossíntese , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Tempo
20.
Stem Cells ; 24(4): 1113-20, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16282439

RESUMO

The identification of stem cell-specific proteins and the elucidation of their novel regulatory pathways may help in the development of protocols for control of their self-renewal and differentiation for cell-based therapies. Nucleostemin is a recently discovered nucleolar protein predominantly associated with proliferating rat neural and embryonic stem cells, and some human cancer cell lines. A comprehensive study of nucleostemin in human adult bone marrow stem cells is lacking. The aim of the study was to determine if nucleostemin is synthesized by adult bone marrow stem cells and to analyze its expression during their expansion and differentiation. Using a multipotential adherent population of stem cells, nucleostemin was localized to the nucleoli and occurred in 43.3% of the cells. There was a high level of expression of nucleostemin mRNA in bone marrow stem cells and this remained unchanged over time during cell expansion in culture. When bone marrow stem cells were stimulated to proliferate by fibroblast growth factor (FGF)-2, nucleostemin expression increased in a dose-dependent manner. Small interfering RNA (siRNA) knockdown of nucleostemin abolished the proliferative effect of FGF-2. When bone marrow stem cells were differentiated into chondrocytes, adipocytes, or osteocytes, nucleostemin expression was 70%-90% lower than in the undifferentiated cells retained in monolayer culture. We conclude that nucleostemin is a marker of undifferentiated human adult bone marrow stem cells and that it is involved in the regulation of proliferation of these cells.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Proteínas de Transporte/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Proteínas Nucleares/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo , Adulto , Sequência de Bases , Biomarcadores/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Diferenciação Celular , Nucléolo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteínas de Ligação ao GTP , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Técnicas In Vitro , Células-Tronco Multipotentes/efeitos dos fármacos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Células Estromais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA