Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
Osteoarthritis Cartilage ; 30(9): 1186-1197, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35487439

RESUMEN

BACKGROUND: Osteoarthritis (OA) affects the entire joint, initially with a low degree of inflammation. Synovitis is correlated with the severity of OA clinical symptoms and cartilage degradation. The synovial lymphatic system (SLS) plays a prominent role in clearing macromolecules within the joint, including the pro-inflammatory cytokines in arthritic status. Scattered evidence shows that impaired SLS drainage function leads to the accumulation of inflammatory factors in the joint and aggravates the progression of OA, and the role of SLS function in OA is less studied. DESIGN: This review summarizes the current understanding of synovial lymphatic function in OA progression and potential regulatory pathways and aims to provide a framework of knowledge for the development of OA treatments targeting lymphatic structure and functions. RESULTS: SLS locates in the subintima layer of the synovium and consists of lymphatic capillaries and lymphatic collecting vessels. Vascular endothelial growth factor C (VEGF-C) is the most critical regulating factor of lymphatic endothelial cells (LECs) and SLS. Nitric oxide production-induced impairment of lymphatic muscle cells (LMCs) and contractile function may attribute to drainage dysfunction. Preclinical evidence suggests that promoting lymphatic drainage may help restore intra-articular homeostasis to attenuate the progression of OA. CONCLUSION: SLS is actively involved in the homeostatic maintenance of the joint. Understanding the drainage function of the SLS at different stages of OA development is essential for further design of therapies targeting the function of these vessels.


Asunto(s)
Vasos Linfáticos , Osteoartritis , Células Endoteliales , Humanos , Sistema Linfático , Osteoartritis/metabolismo , Membrana Sinovial/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo
2.
Osteoarthritis Cartilage ; 25(1): 138-145, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27616684

RESUMEN

OBJECTIVES: The infrapatellar fat pad (IPFP), which is located underneath the patella, close to cartilage surfaces, functions in distributing mechanical load and has been shown to produce cytokines. This study aims to assess the involvement of the IPFP in the progression of post-traumatic osteoarthritis (OA) through investigating the crosstalk between the IPFP and injured cartilage in vitro. METHODS: A single blunt impact (36 MPa) on healthy bovine articular cartilage explants was used to generate traumatized cartilage. Conditioned media from IPFP and traumatized cartilage (FP-CM and TC-CM) were prepared separately. After culturing in FP-CM, the posttraumatic cartilage explants were analyzed for expression of cartilage degeneration associated genes and secretion of the interleukin (IL)-6, into the culture medium. The effect of traumatized cartilage on IPFP was studied by treating IPFP-derived adipocytes and IPFP adipose-derived stromal cells (ADSC) with TC-CM followed by analysis of cytokine expression. RESULTS: FP-CM aggravated glycosaminoglycan (GAG) release in traumatized cartilage, but did not significantly affect healthy cartilage. FP-CM raised gene expression of cyclooxygenase-2, inducible nitric oxide synthase, and IL-6 in traumatized cartilage explants, and lowered expression of tissue inhibitor of metalloproteinases-1, 2, 3, compared to non-conditioned medium. Of particular significance is that medium IL-6 levels increased substantially in both FP-CM and FP-CM treated traumatized cartilage cultures. Extrinsic IL-6 treatment of traumatized cartilage simulated part of the effects of FP-CM. TC-CM elevated levels of IL-6 expression in IPFP derived adipocytes and ADSCs. CONCLUSIONS: IPFP aggravates post-traumatized cartilage degeneration, and IL-6 is a candidate tissue degeneration mediator.


Asunto(s)
Tejido Adiposo/patología , Cartílago Articular/lesiones , Interleucina-6/fisiología , Adipocitos/metabolismo , Animales , Cartílago Articular/metabolismo , Cartílago Articular/patología , Bovinos , Ensayo de Inmunoadsorción Enzimática , Técnicas In Vitro , Osteoartritis/etiología , Rótula/patología , Células del Estroma/metabolismo
3.
Osteoarthritis Cartilage ; 22(8): 1148-57, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24887551

RESUMEN

OBJECTIVE: The long-term performance of cell-seeded matrix-based cartilage constructs depends on (1) the development of sufficient biomechanical properties, and (2) lateral integration with host tissues, both of which require cartilage-specific matrix deposition within the scaffold. In this study, we have examined the potential of tissue-engineered cartilage analogs developed using different cell types, i.e., mesenchymal stem cells (MSCs) vs chondrocytes and de-differentiated chondrocytes, in an established "construct in cartilage ring" model. DESIGN: Cell-laden constructs of differentiated chondrocytes, de-differentiated chondrocytes after two, five or eight population doublings, and MSCs were either implanted into a native cartilage ring immediately after fabrication (immature group) or pre-treated for 21 days in a transforming growth factor-ß3 (TGF-ß3) containing medium prior to implantation. After additional culture for 28 days in a serum-free, chemically defined medium, the extent of lateral integration, and biochemical and biomechanical characteristics of the implants as hybrid constructs were assessed. RESULTS: The quality of integration, the amount of accumulated cartilage-specific matrix components and associated biomechanical properties were found to be highest when using differentiated chondrocytes. De-differentiation of chondrocytes negatively impacted the properties of the implants, as even two population doublings of the chondrocytes in culture significantly lowered cartilage repair capacity. In contrast, MSCs showed chondrogenic differentiation with TGF-ß3 pre-treatment and superior integrational behavior. CONCLUSIONS: Chondrocyte expansion and de-differentiation impaired the cell response, resulting in inferior cartilage repair in vitro. With TGF-ß3 pre-treatment, MSCs were able to undergo sustained chondrogenic differentiation and exhibited superior matrix deposition and integration compared to de-differentiated chondrocytes.


Asunto(s)
Cartílago Articular , Condrocitos , Células Madre Mesenquimatosas , Ingeniería de Tejidos/métodos , Animales , Bovinos , Diferenciación Celular , Condrogénesis , Regeneración Tisular Dirigida/métodos , Hidrogel de Polietilenoglicol-Dimetacrilato , Técnicas In Vitro , Factor de Crecimiento Transformador beta3
4.
Osteoarthritis Cartilage ; 19(2): 213-21, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21087676

RESUMEN

OBJECTIVE: Cell-based tissue engineering strategies are currently in clinical use and continue to be developed at a rapid pace for the repair of cartilage defects. Regardless of the repair methodology, chondrocytes within newly regenerated cartilage remain susceptible to the abnormal inflammatory and mechanical environments that underlie osteoarthritic disease, likely compromising the implant's integration, function, and longevity. The present study investigates the use of parathyroid hormone-related peptide (PTHrP) overexpression for chondroprotection. DESIGN: Bovine articular chondrocytes were transfected with human PTHrP (hPTHrP) constructs (1-141 or 1-173) and subjected to injurious cyclic tensile strain (CTS; 0.5 Hz and 16% elongation) for 48 h. mRNA expression of matrix remodeling, inflammatory signaling, hypertrophic, and apoptotic genes were examined with real-time reverse transcription polymerase chain reaction. Nitric oxide (NO) and prostaglandin E2 (PGE2) production were measured using the Griess assay and enzyme immunoassay (EIA), respectively. RESULTS: CTS-induced an arthritic phenotype in articular chondrocytes as indicated by increased gene expression of collagenases and aggrecanases and increased production of NO and PGE2. Additionally, CTS increased collagen type X (Col10a1) mRNA expression, whereas overexpression of either hPTHrP isoform inhibited CTS-induced Col10a1 gene expression. However, hPTHrP 1-141 augmented CTS-induced NO and PGE2 production, and neither hPTHrP isoform had any significant effect on apoptotic genes. CONCLUSIONS: Our results suggest that chondrocytes overexpressing PTHrP resist mechanical strain-induced hypertrophic-like changes. Therapeutic PTHrP gene transfer may be considered for chondroprotection applications in newly regenerated cartilage.


Asunto(s)
Artritis Experimental/metabolismo , Condrocitos/fisiología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Estrés Mecánico , Animales , Artritis Experimental/genética , Cartílago Articular/citología , Bovinos , Células Cultivadas , Dinoprostona/metabolismo , Regulación de la Expresión Génica , Óxido Nítrico/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/genética , Fenotipo , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/metabolismo , Transfección
5.
J Tissue Eng Regen Med ; 4(1): 25-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19834956

RESUMEN

The presence of a defect in mature articular cartilage can lead to degenerative changes of the joint. This is in part caused by the inability of cartilage to regenerate tissue that is capable of spanning a fissure or crack. In this study, we hypothesized that introduction of a biodegradable cell-seeded nanofibrous hydrogel, Puramatrix(), into a cartilage gap would facilitate the generation of a mechanically stable interface. The effects of chondrocyte incorporation within the hydrogel and supplementation with transforming growth factor-beta3 (TGFbeta3), a known regulator of cell growth and differentiation, on cartilage integration were examined mechanically and histologically as a function of cell density and incubation time. When supplemented with TGFbeta3, the cell-seeded hydrogel exhibited abundant matrix generation within the hydrogel and a corresponding increase in maximum push-out stress as compared to all other groups. Furthermore, initial cell seeding density affected interfacial strength in a time-dependent manner. This study suggests that a cell-seeded TGFbeta3-supplemented hydrogel can encourage integration between two opposing pieces of articular cartilage.


Asunto(s)
Cartílago Articular/fisiología , Ingeniería de Tejidos/métodos , Animales , Fenómenos Biomecánicos , Cartílago Articular/efectos de los fármacos , Cartílago Articular/lesiones , Bovinos , Supervivencia Celular , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/fisiología , Condrocitos/trasplante , Hidrogeles , Modelos Biológicos , Nanofibras , Regeneración/efectos de los fármacos , Regeneración/fisiología , Factor de Crecimiento Transformador beta3/farmacología
6.
Oncogene ; 28(38): 3401-11, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19597468

RESUMEN

The development of pulmonary metastasis is the major cause of death in osteosarcoma, and its molecular basis is poorly understood. In this study, we show that beta4 integrin is highly expressed in human osteosarcoma cell lines and tumor samples. Furthermore, highly metastatic MNNG-HOS cells have increased levels of beta4 integrin. Suppression of beta4 integrin expression by shRNA and disruption of beta4 integrin function by transfection of dominant-negative beta4 integrin was sufficient to revert this highly metastatic phenotype in the MNNG-HOS model without significantly affecting primary tumor growth. These findings suggest a role for beta4 integrin expression in the metastatic phenotype in human osteosarcoma cells. In addition, we identified a previously uncharacterized interaction between beta4 integrin and ezrin, a membrane-cytoskeletal linker protein that is implicated in the metastatic behavior of osteosarcoma. The beta4 integrin-ezrin interaction appears to be critical for maintenance of beta4 integrin expression. These data begin to integrate ezrin and beta4 integrin expression into a model of action for the mechanism of osteosarcoma metastases.


Asunto(s)
Neoplasias Óseas/patología , Proteínas del Citoesqueleto/fisiología , Integrina beta4/fisiología , Osteosarcoma/secundario , Línea Celular Tumoral , Humanos , Integrina beta4/análisis
7.
J Tissue Eng Regen Med ; 3(2): 129-38, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19170141

RESUMEN

Mesenchymal stem cells (MSCs) derived from adult tissues are an important candidate cell type for cell-based tissue engineering and regenerative medicine. Currently, clinical applications for MSCs require additional surgical procedures to harvest the autologous MSCs (i.e. from bone marrow) or commercial allogeneic alternatives. We have recently identified a population of mesenchymal progenitor cells (MPCs) in traumatized muscle tissue that has been surgically debrided from traumatic orthopaedic extremity wounds. The purpose of this study was to evaluate whether MPCs derived from traumatized muscle may provide a clinical alternative to bone-marrow MSCs, by comparing their morphology, proliferation capacity, cell surface epitope profile and differentiation capacity. After digesting the muscle tissue with collagenase, the MPCs were enriched by a direct plating technique. The morphology and proliferation rate of the muscle-derived MPCs was similar to bone-marrow derived MSCs. Both populations expressed cell surface markers characteristic for MSCs (CD 73, CD 90 and CD105), and did not express markers typically absent on MSCs (CD14, CD34 and CD45). After 21 days in specific differentiation media, the histological staining and gene expression of the MPCs and MSCs was characteristic for differentiation into osteoblasts, chondrocytes and adipocytes, but not into myoblasts. Our findings demonstrate that traumatized muscle-derived MPCs exhibit a similar phenotype and resemble MSCs derived from the bone marrow. MPCs harvested from traumatized muscle tissue may be considered for applications in tissue engineering and regenerative medicine following orthopaedic trauma requiring circumferential debridement.


Asunto(s)
Células Madre Mesenquimatosas/citología , Músculos/patología , Antígenos CD/inmunología , Secuencia de Bases , Diferenciación Celular , Proliferación Celular , Cartilla de ADN , Epítopos/inmunología , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunofenotipificación , Células Madre Mesenquimatosas/inmunología , Músculos/lesiones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ingeniería de Tejidos
8.
Osteoarthritis Cartilage ; 15(9): 1025-33, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17498976

RESUMEN

OBJECTIVE: To determine whether the functional properties of tissue-engineered constructs cultured in a chemically-defined medium supplemented briefly with TGF-beta3 can be enhanced with the application of dynamic deformational loading. METHODS: Primary immature bovine cells (2-3 months old) were encapsulated in agarose hydrogel (2%, 30 x 10(6)cells/ml) and cultured in chemically-defined medium supplemented for the first 2 weeks with transforming growth factor beta 3 (TGF-beta3) (10 microg/ml). Physiologic deformational loading (1 Hz, 3 h/day, 10% unconfined deformation initially and tapering to 2% peak-to-peak deformation by day 42) was applied either concurrent with or after the period of TGF-beta3 supplementation. Mechanical and biochemical properties were evaluated up to day 56. RESULTS: Dynamic deformational loading applied concurrently with TGF-beta3 supplementation yielded significantly lower (-90%) overall mechanical properties when compared to free-swelling controls. In contrast, the same loading protocol applied after the discontinuation of the growth factor resulted in significantly increased (+10%) overall mechanical properties relative to free-swelling controls. Equilibrium modulus values reach 1306+/-79 kPa and glycosaminoglycan levels reach 8.7+/-1.6% w.w. during this 8-week period and are similar to host cartilage properties (994+/-280 kPa, 6.3+/-0.9% w.w.). CONCLUSIONS: An optimal strategy for the functional tissue engineering of articular cartilage, particularly to accelerate construct development, may incorporate sequential application of different growth factors and applied deformational loading.


Asunto(s)
Cartílago Articular/citología , Condrocitos/citología , Estrés Mecánico , Ingeniería de Tejidos/métodos , Animales , Cartílago Articular/metabolismo , Cartílago Articular/fisiología , Bovinos , Técnicas de Cultivo de Célula , Condrocitos/fisiología , Colágeno/análisis , Glicosaminoglicanos/análisis , Modelos Biológicos , Factor de Crecimiento Transformador beta3
9.
Osteoarthritis Cartilage ; 15(6): 646-55, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17337215

RESUMEN

OBJECTIVES: Glucosamine (GlcN), a natural amino monosaccharide, is a constituent of glycosaminoglycans (GAGs) found in hyaline cartilage. GlcN salts constitute a new class of nutraceutical components with putative chondroprotective activity, which may target chondrocytes as well as chondroprogenitors cells, such as mesenchymal stem cells (MSCs), during cartilage turnover and repair. In the present study, we examined the effects of GlcN on chondrogenesis of human MSCs (hMSCs) and the phenotype of normal and osteoarthritic human articular chondrocytes, using an in vitro pellet culture model maintained in a defined medium. METHODS: hMSCs and normal and osteoarthritic human chondrocytes grown as pellet cultures, stimulated or not with interleukin-1beta (IL-1beta), were treated with varying doses of GlcN. Expression of cartilage matrix genes and cartilage degrading enzymes was determined by semiquantitative and quantitative real-time reverse transcription polymerase chain reaction (RT-PCR), and by histological staining of cartilage markers, as well as sulfated GAG (sGAG) analysis and Western blotting. RESULTS: Chondrocytes grown in the presence of serum for 11 days showed decreased expression of the cartilage matrix genes, collagen type II (collagen II) and aggrecan, as early as day 3, which was reversed with GlcN treatment by day 11. Both hMSCs and chondrocytes grown as pellet cultures in defined medium and treated with 100 microM GlcN exhibited enhanced expression of collagen II and aggrecan as well as increased content of sGAG, when compared to control untreated pellets. However, high doses of GlcN (10-20mM) were inhibitory. GlcN treatment partially blocked IL-1beta mediated downregulation of collagen II and aggrecan expression and inhibited expression of the matrix degrading enzyme, matrix metalloproteinase 13 (MMP-13), in both chondrocytes and hMSCs undergoing chondrogenesis. CONCLUSIONS: These observations suggest that GlcN treatment enhances hMSC chondrogenesis and maintains cartilage matrix gene expression in chondrocytes, which may account for some of the reported chondroprotective properties of GlcN on cartilage.


Asunto(s)
Cartílago Articular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glucosamina/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoartritis/metabolismo , Cartílago Articular/citología , Cartílago Articular/metabolismo , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Humanos , Metaloproteinasa 13 de la Matriz/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteoartritis/patología
10.
Biomech Model Mechanobiol ; 6(1-2): 113-25, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16691412

RESUMEN

This study explored the biologic response of chondrocytes and mesenchymal stem cells (MSCs) to a dynamic mechanical loading regime. We developed a time-efficient methodology for monitoring regional changes in extracellular matrix gene transcription using reporter promoter constructs. Specifically, transfected cells were homogenously distributed throughout agarose hydrogel constructs, and spatial and temporal gene expression and the ability to form functional ECM were analyzed in response to dynamic mechanical stimuli. Theoretical analyses were used to predict the physical signals generated within the gel in response to these loading regimes. Using a custom compression bioreactor system, changes in aggrecan and type II collagen promoter activity in transfected chondrocyte-laden cylindrical constructs were evaluated in response to a range of loading frequencies and durations. In general, aggrecan promoter activity increased with increasing duration of loading, particularly in the outer annulus region. Interestingly, type II collagen promoter activity decreased in this annular region under identical loading conditions. In addition, we explored the role of mechanical compression in directing chondrogenic differentiation of MSCs by monitoring short-term aggrecan promoter activity. As an example of long-term utility, a specific loading protocol was applied to MSC-laden constructs for 5 days, and the resultant changes in glycosaminoglycan (GAG) production were evaluated over a 4-week period. This dynamic loading regime increased not only short-term aggrecan transcriptional activity but also GAG deposition in long-term culture. These results demonstrate the utility of a new reporter promoter system for optimizing loading protocols to improve the outcome of engineered chondrocyte- and MSC-laden cartilaginous constructs.


Asunto(s)
Cartílago/metabolismo , Condrocitos/metabolismo , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Transcripción Genética , Soporte de Peso/fisiología , Agrecanos/genética , Animales , Reactores Biológicos , Bovinos , Técnicas de Cultivo de Célula , Condrocitos/citología , Colágeno Tipo II/genética , Fuerza Compresiva , Análisis de Elementos Finitos , Geles , Genes Reporteros , Glicosaminoglicanos/metabolismo , Luciferasas de Renilla/metabolismo , Células Madre Mesenquimatosas/citología , Regiones Promotoras Genéticas/genética
11.
Osteoarthritis Cartilage ; 14(2): 179-89, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16257243

RESUMEN

BACKGROUND: The developmental history of the chondrocyte results in a cell whose biosynthetic activities are optimized to maintain the concentration and organization of a mechanically functional cartilaginous extracellular matrix. While useful for cartilage tissue engineering studies, the limited supply of healthy autologous chondrocytes may preclude their clinical use. Consequently, multipotential mesenchymal stem cells (MSCs) have been proposed as an alternative cell source. OBJECTIVE: While MSCs undergo chondrogenesis, few studies have assessed the mechanical integrity of their forming matrix. Furthermore, efficiency of matrix formation must be determined in comparison to healthy chondrocytes from the same donor. Given the scarcity of healthy human tissue, this study determined the feasibility of isolating bovine chondrocytes and MSCs, and examined their long-term maturation in three-dimensional agarose culture. EXPERIMENTAL DESIGN: Bovine MSCs were seeded in agarose and induced to undergo chondrogenesis. Mechanical and biochemical properties of MSC-laden constructs were monitored over a 10-week period and compared to those of chondrocytes derived from the same group of animals maintained similarly. RESULTS: Our results show that while chondrogenesis does occur in MSC-laden hydrogels, the amount of the forming matrix and measures of its mechanical properties are lower than that produced by chondrocytes under the same conditions. Furthermore, some important properties, particularly glycosaminoglycan content and equilibrium modulus, plateau with time in MSC-laden constructs, suggesting that diminished capacity is not the result of delayed differentiation. CONCLUSIONS: These findings suggest that while MSCs do generate constructs with substantial cartilaginous properties, further optimization must be done to achieve levels similar to those produced by chondrocytes.


Asunto(s)
Condrocitos/citología , Condrogénesis , Células Madre Mesenquimatosas/citología , Animales , Fenómenos Biomecánicos , Bovinos , Técnicas de Cultivo de Célula , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Condrocitos/metabolismo , Colágeno/análisis , Medios de Cultivo , ADN/análisis , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestructura , Células Madre Mesenquimatosas/metabolismo , Proteoglicanos/análisis , Proteoglicanos/metabolismo , Sefarosa , Factores de Tiempo
12.
Biomaterials ; 26(6): 599-609, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15282138

RESUMEN

The utilization of adult stem cells in tissue engineering is a promising solution to the problem of tissue or organ shortage. Adult bone marrow derived mesenchymal stem cells (MSCs) are undifferentiated, multipotential cells which are capable of giving rise to chondrocytes when maintained in a three-dimensional culture and treated with members of the transforming growth factor-beta (TGF-beta) family of growth factors. In this study, we fabricated a nanofibrous scaffold (NFS) made of a synthetic biodegradable polymer, poly(-caprolactone) (PCL), and examined its ability to support in vitro chondrogenesis of MSCs. The electrospun PCL porous scaffold was constructed of uniform, randomly oriented nanofibers with a diameter of 700 nm, and structural integrity of this scaffold was maintained over a 21-day culture period. MSCs cultured in NFSs in the presence of TGF-beta1 differentiated to a chondrocytic phenotype, as evidenced by chondrocyte-specific gene expression and synthesis of cartilage-associated extracellular matrix (ECM) proteins. The level of chondrogenesis observed in MSCs seeded within NFSs was comparable to that observed for MSCs maintained as cell aggregates or pellets, a widely used culture protocol for studying chondrogenesis of MSCs in vitro. Due to the physical nature and improved mechanical properties of NFSs, particularly in comparison to cell pellets, the findings reported here suggest that the PCL NFS is a practical carrier for MSC transplantation, and represents a candidate scaffold for cell-based tissue engineering approaches to cartilage repair.


Asunto(s)
Cartílago/citología , Técnicas de Cultivo de Célula/instrumentación , Condrocitos/citología , Células Madre Mesenquimatosas/citología , Nanoestructuras , Ingeniería de Tejidos/instrumentación , Anciano , Biodegradación Ambiental , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas/citología , Células Cultivadas/efectos de los fármacos , Condrocitos/efectos de los fármacos , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/genética , Glicosaminoglicanos/biosíntesis , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Microscopía Electrónica de Rastreo , Persona de Mediana Edad , ARN Mensajero/biosíntesis , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1
13.
J Cell Mol Med ; 8(3): 301-16, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15491506

RESUMEN

A considerable amount of retrospective data is available that describes putative mesenchymal stem cells (MSCs). However, there is still very little knowledge available that documents the properties of a MSC in its native environment. Although the precise identity of MSCs remains a challenge, further understanding of their biological properties will be greatly advanced by analyzing the mechanisms that govern their self-renewal and differentiation potential. This review begins with the current state of knowledge on the biology of MSCs, specifically with respect to their existence in the adult organism and postulation of their biological niche. While MSCs are considered suitable candidates for cell-based strategies owing to their intrinsic capacity to self-renew and differentiate, there is currently little information available regarding the molecular mechanisms that govern their stem cell potential. We propose here a model for the regulation of MSC differentiation, and recent findings regarding the regulation of MSC differentiation are discussed. Current research efforts focused on elucidating the mechanisms regulating MSC differentiation should facilitate the design of optimal in vitro culture conditions to enhance their clinical utility cell and gene therapy.


Asunto(s)
Linaje de la Célula , Terapia Genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células del Estroma/citología , Diferenciación Celular , Humanos , Osteogénesis
14.
Cytotherapy ; 6(6): 596-601, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15773023

RESUMEN

Cartilage repair and regeneration by stem cell-based tissue engineering could be of enormous therapeutic and economic potential benefit for an aging population. However, to use stem cells effectively, their natural environment must be understood in order to expand them in vitro without compromising their multilineage potential and their specific differentiation program. Collaboration between diverse academic disciplines and between research and regulatory government agencies and industry is crucial before cell-based cartilage tissue engineering can achieve its full therapeutic potential.


Asunto(s)
Cartílago/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Ingeniería de Tejidos , Adulto , Materiales Biocompatibles , Condrogénesis , Humanos , Regeneración
15.
J Cell Biochem ; 89(4): 755-70, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12858341

RESUMEN

During development, calcium (Ca) is actively transported by placental trophoblasts to meet fetal nutritional and the skeletal mineralization needs. Maternal exposure to estrogenic pesticides, such as 1,1-bis(p-chlorophenyl)-2,2,2-trichloroethane (DDT) and methoxychlor (MTC), has been shown to result in reproductive disorders and/or abnormal fetal development. In this study, we have examined the effects of exposure of trophoblastic cells to MTC and DTT, in comparison to 17beta-estradiol (E2) and diethylstilbestrol (DES), to test the hypothesis that cellular Ca handling is a target for these endocrine disruptive components. Treatment with DDT, MTC, DES, or E2 increased cellular Ca uptake, and the expression of trophoblast-specific human Ca binding protein (HCaBP) was down-regulated by both MTC and DDT. Treatment with MTC, DDT, and DES inhibited cell proliferation, induced apoptosis, and suppressed expression of several trophoblast differentiation marker genes. These effects were reversed by overexpression of metallothionein IIa, a gene highly responsive to cadmium and other metals. These results strongly suggest that trophoblast Ca handling functions are endocrinally modulated, and that their alteration by candidate endocrine disruptors, such as MTC and DDT, constitutes a possible pathway of the harmful effects of these components on fetal development.


Asunto(s)
Calcio/metabolismo , DDT/efectos adversos , Dietilestilbestrol/efectos adversos , Estradiol/efectos adversos , Metoxicloro/efectos adversos , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Adenosina Trifosfatasas/metabolismo , Proteínas de Unión al Calcio/biosíntesis , Proteínas de Unión al Calcio/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Regulación hacia Abajo , Activación Enzimática/efectos de los fármacos , Estradiol/análogos & derivados , Marcadores Genéticos , Humanos , Metalotioneína/metabolismo , Metalotioneína/farmacología , Receptores de Estrógenos/análisis , Receptores de Estrógenos/biosíntesis , Receptores de Progesterona/análisis , Receptores de Progesterona/biosíntesis , Trofoblastos/citología
16.
Osteoarthritis Cartilage ; 11(6): 442-54, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12801484

RESUMEN

OBJECTIVE: Cartilage oligomeric matrix protein (COMP) mutations have been identified as responsible for two arthritic disorders, multiple epiphyseal dysplasia (MED) and pseudoachondroplasia (PSACH). However, the function of COMP in chondrogenic differentiation is largely unknown. Our investigation focuses on analyzing the function of normal COMP protein in cartilage biology. METHODS AND RESULTS: To explore the function of COMP we make use of an in vitro model system for chondrogenesis, consisting of murine C3H10T1/2 mesenchymal cells maintained as a high-density micromass culture and stimulated with bone morphogenetic protein 2 (BMP-2). Under these culture conditions, C3H10T1/2 cells undergo active chondrogenesis in a manner analogous to that of embryonic limb mesenchymal cells, and have been shown to serve as a valid model system to investigate the mechanisms regulating mesenchymal chondrogenesis. Our results indicate that ectopic COMP expression enhances several early aspects of chondrogenesis induced by BMP-2 in this system, indicating that COMP functions in part to positively regulate chondrogenesis. Additionally, COMP has inhibitory effects on proliferation of cells in monolayer. However, at later times in micromass culture, ectopic COMP expression in the presence of BMP-2 causes an increase in apoptosis, with an accompanying reduction in cell numbers in the micromass culture. However, the remaining cells retain their chondrogenic phenotype. CONCLUSIONS: These data suggest that COMP and BMP-2 signaling converge to regulate the fate of these cells in vitro by affecting both early and late stages of chondrogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Condrogénesis/fisiología , Proteínas de la Matriz Extracelular/fisiología , Glicoproteínas/fisiología , Mesodermo/fisiología , Factor de Crecimiento Transformador beta , Animales , Northern Blotting , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/fisiología , Células Cultivadas , Electroforesis en Gel de Poliacrilamida/métodos , Hibridación in Situ , Proteínas Matrilinas , Ratones , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
17.
Biomaterials ; 24(7): 1213-21, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12527262

RESUMEN

The nature of the extracellular matrix (ECM) is crucial in regulating cell functions via cell-matrix interactions, cytoskeletal organization, and integrin-mediated signaling. In bone, the ECM is composed of proteins such as collagen (CO), fibronectin (FN), laminin (LM), vitronectin (VN), osteopontin (OP) and osteonectin (ON). For bone tissue engineering, the ECM should also be considered in terms of its function in mediating cell adhesion to biomaterials. This study examined ECM production, cytoskeletal organization, and adhesion of primary human osteoblastic cells on biodegradable matrices applicable for tissue engineering, namely polylactic-co-glycolic acid 50:50 (PLAGA) and polylactic acid (PLA). We hypothesized that the osteocompatible, biodegradable polymer surfaces promote the production of bone-specific ECM proteins in a manner dependent on polymer composition. We first examined whether the PLAGA and PLA matrices could support human osteoblastic cell growth by measuring cell adhesion at 3, 6 and 12h post-plating. Adhesion on PLAGA was consistently higher than on PLA throughout the duration of the experiment, and comparable to tissue culture polystyrene (TCPS). ECM components, including CO, FN, LM, ON, OP and VN, produced on the surface of the polymers were quantified by ELISA and localized by immunofluorescence staining. All of these proteins were present at significantly higher levels on PLAGA compared to PLA or TCPS surfaces. On PLAGA, OP and ON were the most abundant ECM components, followed by CO, FN, VN and LN. Immunofluorescence revealed an extracellular distribution for CO and FN, whereas OP and ON were found both intracellularly as well as extracellularly on the polymer. In addition, the actin cytoskeletal network was more extensive in osteoblasts cultured on PLAGA than on PLA or TCPS. In summary, we found that osteoblasts plated on PLAGA adhered better to the substrate, produced higher levels of ECM molecules, and showed greater cytoskeletal organization than on PLA and TCPS. We propose that this difference in ECM composition is functionally related to the enhanced cell adhesion observed on PLAGA. There is initial evidence that specific composition of the PLAGA polymer favors the ECM. Future studies will seek to optimize ECM production on these matrices for bone tissue engineering applications.


Asunto(s)
Adhesión Celular/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Osteoblastos/metabolismo , Polímeros/metabolismo , Ingeniería de Tejidos , Biodegradación Ambiental , Huesos/citología , Células Cultivadas , Medios de Cultivo/química , Humanos , Osteoblastos/química , Osteoblastos/citología , Polímeros/química
18.
Osteoarthritis Cartilage ; 10(8): 638-45, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12479386

RESUMEN

OBJECTIVE: A subgroup of patients with pseudoachondroplasia (PSACH) and multiple epiphyseal dysplasia (MED) have been found to harbor mutations within the cartilage oligomeric matrix protein (COMP) gene. These two diseases are autosomal dominant disorders that are characterized by an early onset of osteoarthritis (OA). The COMP gene is expressed primarily in chondrocytes in articular cartilage as well as in tendon and ligament. Therefore, control over tissue specific COMP expression may be an important aspect in cartilage biology. To begin an analysis of the regulation of COMP expression, we have cloned, sequenced and characterized the entire genomic clone for mouse COMP that includes the COMP promoter. METHODS AND RESULTS: The COMP coding region spans 19 exons over approximately 8.4 kb of DNA. The arrangement and size of the exons have a remarkable similarity to those of the human COMP genomic sequence, indicating a significant degree of genomic conservation. Analysis of a 453 basepair region of the putative COMP promoter reveals two strong transcriptional repressor elements located between position -356 and -304 and between -251 and -180, relative to the start site for transcription. These repressor elements down-regulate transcription from the promoter in a broad spectrum of cell lines. Removal of the repressor DNA sequence from the COMP promoter leads to significant enhancement in transcriptional activity, indicating that this region acts in a dominant manner to transcriptional activators located more proximal to the start site of transcription. This region also represses transcription when linked to a heterologous promoter. CONCLUSIONS: This repressor region probably down-regulates transcription from the COMP promoter in vivo. It may help to repress transcription of COMP in non-cartilaginous tissues and/or may aid in the expression of COMP to the appropriate level in tissues such as cartilage, tendon and ligament.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Glicoproteínas/genética , Osteoartritis/genética , Regiones Promotoras Genéticas/genética , Proteínas Represoras/genética , Transcripción Genética/genética , Acondroplasia/genética , Animales , Secuencia de Bases/genética , Proteína de la Matriz Oligomérica del Cartílago , Cartílago Articular/fisiología , Línea Celular , Células Cultivadas , Regulación hacia Abajo/genética , Humanos , Proteínas Matrilinas , Ratones , Datos de Secuencia Molecular , Mutación , Osteocondrodisplasias/genética , Ratas , Transfección
19.
Calcif Tissue Int ; 71(5): 447-58, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12232673

RESUMEN

We have established a new adult human bone marrow-derived cell line hMPC 32F, stably transduced with human papilloma virus type 16 E6/E7 genes, that displays mesenchymal multilineage differentiation ability in vitro. The hMPC 32F cells exhibited a population doubling time of 22 h and have been maintained in culture for about 20 passages. When cultured in conditions promoting osteogenic, adipogenic, or chondrogenic differentiation, hMPC 32F cells expressed mature differentiated phenotypes. These include (1) osteoblastic phenotype characterized by upregulated alkaline phosphatase (ALP) expression and extracellular matrix mineralization, (2) adipocytic phenotype with the presence of intracellular lipid droplets, and (3) chondrocytic phenotype of round cells surrounded by a sulfated proteoglycan-rich matrix. In addition, the hMPC 32F cells expressed differentiation lineage-specific genes, as detected by RT-PCR. Furthermore, osteogenic and adipogenic cultures responded to regulatory factors such as transforming growth factor-beta1 (TGF-beta1) and 1alpha, 25-dihydroxyvitamin D3 (1,25(OH)2D3). Thus, continuous treatment of osteogenic cultures for 2 weeks with TGF-beta1 decreased ALP activity and mRNA expression and inhibited osteocalcin mRNA expression and matrix mineralization, whereas l,25(OH)2D3 had an additive, stimulatory effect. In adipogenic cultures, treatment with TGF-beta1 for 2 weeks markedly inhibited adipogenesis whereas 1,25(OH)2D3 had no obvious effect. Finally, clonal analysis of hMPC 32F cells revealed a high percentage of multipotent clones, although clones of more restricted differentiation potential were also present. These characteristics of the hMPC 32F cell line suggest their pluripotent, progenitor, and nontransformed nature and indicate their potential application for studying the mechanisms governing developmental potential of adult human bone marrow mesenchymal progenitor cells.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Genes Virales , Papillomaviridae/genética , Células Madre/citología , Adipocitos/citología , Adipocitos/metabolismo , Adulto , Células de la Médula Ósea/fisiología , Linaje de la Célula , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Femenino , Humanos , Osteoblastos/citología , Osteoblastos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/fisiología , Transducción Genética
20.
Ann N Y Acad Sci ; 961: 134-8, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12081882

RESUMEN

Marrow stroma-derived cells (MSC) are highly proliferative, multipotential cells that have been considered as ideal candidate cells for autologous tissue engineering applications. In this study, we have characterized the chondrogenic potential of human MSCs in both a PLA/alginate amalgam and pure PLA macrostructure as model three-dimensional constructs to support both chondrogenic differentiation and proliferation following TGF-beta treatment. MSCs were seeded in experimental groups that consisted of PLA-loaded constructs and PLA/alginate amalgams with and without recombinant human TGF-beta1. Chondrogenesis of the PLA and the PLA/alginate amalgam cultures was assessed at weekly intervals by histology, immunohistochemistry, scanning electron microscopy, sulfate incorporation, and RT-PCR. Chondrogenic differentiation occurs within a polymeric macrostructure with TGF-beta1 treatment as indicated by histological, immunohistochemical, sulfate incorporation, and gene expression profiles. This macrostructure can be further encased in an alginate gel/solution to optimize cell shape and to confine growth factors and cells within the polymer construct, while the polymeric scaffold provides appropriate mechanical/tissue support. The stable three-dimensional PLA/alginate amalgam represents a novel candidate system of mesenchymal chondrogenesis, which is amendable to investigation of mechanical and biological factors that normally modulate cartilage development and formation as well as a potential tissue engineering construct for cartilage repair.


Asunto(s)
Alginatos/farmacología , Materiales Biocompatibles/farmacología , Cartílago/metabolismo , Polímeros/farmacología , Ingeniería de Tejidos , Alginatos/metabolismo , Células de la Médula Ósea/citología , Diferenciación Celular , Células Cultivadas , Condrocitos/metabolismo , Humanos , Inmunohistoquímica , Microscopía Electrónica de Rastreo , Polímeros/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/citología , Sulfatos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA