Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Stem Cells ; 38(11): 1438-1453, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32652878

RESUMEN

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.


Asunto(s)
Metaloproteinasa 13 de la Matriz/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Osteoartritis/terapia , Ingeniería de Tejidos/métodos , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo
2.
Stem Cells ; 35(11): 2280-2291, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28833807

RESUMEN

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.


Asunto(s)
Condrogénesis/genética , Células Madre Mesenquimatosas/metabolismo , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Proteína Wnt-5a/genética , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Ovinos , Ingeniería de Tejidos , Proteína Wnt-5a/metabolismo
3.
Biomacromolecules ; 14(5): 1287-98, 2013 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-23534615

RESUMEN

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.


Asunto(s)
Materiales Biocompatibles/farmacología , Celulosa/química , Condrocitos/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Seda/química , Ingeniería de Tejidos/métodos , Agrecanos/genética , Agrecanos/metabolismo , Materiales Biocompatibles/química , Diferenciación Celular , Condrocitos/citología , Condrocitos/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Líquidos Iónicos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Resistencia a la Tracción , Andamios del Tejido
4.
Nat Rev Rheumatol ; 19(7): 403-416, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37296196

RESUMEN

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.


Asunto(s)
Cartílago Articular , Osteoartritis , Humanos , Condrocitos , Osteoartritis/terapia , Regeneración
5.
Biomaterials ; 285: 121547, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35533445

RESUMEN

Targeting stem cells to cartilage lesions has the potential to enhance engraftment and chondrogenesis. Denatured type II collagen fibrils (gelatin) are exposed in lesions at the surface of osteoarthritic articular cartilage and are therefore ideal target sites. We have designed and investigated chimeric mutants of the three modules of the MMP-2 collagen binding domain (CBD) as potential ligands for stem cell targeting. We expressed full-length CBD for the first time and used it to identify the most important amino acid residues for binding to gelatin. Module 2 of CBD had the highest affinity binding to both Type I and Type II gelatin, whereas module 1 showed specificity for type II gelatin and module 3 for type I gelatin. We went on to generate chimeric forms of CBD consisting of three repeats of module 1 (111), module 2 (222) or module 3 (333). 111 lacked solubility and could not be further characterised. However 222 was found to bind to type II gelatin 14 times better than CBD, suggesting it would be optimal for attachment to cartilage lesions, whilst 333 was found to bind to type I gelatin 12 times better than CBD, suggesting it would be optimal for attachment to lesions in type I collagen-rich tissues. We coated 222 onto the external membrane of Mesenchymal Stem Cells and demonstrated higher attachment of the coated cells to type II gelatin than uncoated cells. We conclude that the three modules of CBD each have specific biological properties that can be exploited for targeting stem cells to cartilage lesions and other pathological sites.


Asunto(s)
Cartílago Articular , Metaloproteinasa 2 de la Matriz , Proteínas Portadoras/metabolismo , Cartílago/metabolismo , Cartílago Articular/metabolismo , Colágeno Tipo I/metabolismo , Gelatina , Metaloproteinasa 2 de la Matriz/metabolismo , Membranas Artificiales , Unión Proteica , Estructura Terciaria de Proteína , Células Madre/metabolismo
6.
Stem Cells ; 28(11): 1992-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20882533

RESUMEN

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.


Asunto(s)
Cartílago/citología , Regeneración/fisiología , Células Madre Adultas/citología , Animales , Condrogénesis/fisiología , Humanos , Células Madre Mesenquimatosas/citología , Modelos Biológicos
7.
Lancet ; 372(9655): 2023-30, 2008 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-19022496

RESUMEN

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.


Asunto(s)
Broncomalacia/fisiopatología , Condrocitos/citología , Ingeniería de Tejidos/métodos , Tráquea/trasplante , Adulto , Broncomalacia/terapia , Cadáver , Femenino , Humanos , Periodo Posoperatorio , Pruebas de Función Respiratoria , Tráquea/citología
8.
Matrix Biol ; 27(6): 526-34, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18534835

RESUMEN

Lumican is a glycoprotein that is found in the extracellular matrix of many connective tissues, including cartilage. It is a member of the small leucine-rich repeat proteoglycans family and along with two others, decorin and fibromodulin, has the capacity to bind to fibrillar collagens and limit their growth. Cartilage tissue engineering provides a potential method for the production of three-dimensional tissue for implantation into eroded joints. Many studies have demonstrated the growth of cartilage in vitro. However in all cases, biochemical analysis of the tissue revealed a significant deficit in the collagen content. We have now tested the hypothesis that the reduced collagen accumulation in engineered cartilage is a result of over-expression of decorin, fibromodulin or lumican. We have found that the lumican gene and protein are both over-expressed in engineered compared to natural cartilage whereas this is not the case for decorin or fibromodulin. Using a small hairpin lumican antisense sequence we were able to knockdown the lumican gene and protein expression in chondrocytes being used for tissue engineering. This resulted in increased accumulation of type II collagen (the major collagen of cartilage) whilst there was no significant alteration in the proteoglycan content. Furthermore, the antisense knockdown of lumican resulted in an increase in the average collagen fibril diameter measured by transmission electron microscopy. These results suggest that lumican plays a pivotal role in the development of tissue engineered cartilage and that regulation of this protein may be important for the production of high-quality implants.


Asunto(s)
Cartílago , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Colágeno/metabolismo , Sulfato de Queratano/metabolismo , Ingeniería de Tejidos , Animales , Cartílago/citología , Cartílago/metabolismo , Bovinos , Proteoglicanos Tipo Condroitín Sulfato/genética , Colágeno/ultraestructura , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Eliminación de Gen , Humanos , Sulfato de Queratano/genética , Lumican , Ratones , Proteoglicanos/genética , Proteoglicanos/metabolismo , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Distribución Aleatoria
9.
Stem Cells ; 25(10): 2460-8, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17615267

RESUMEN

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.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Condrocitos/citología , Condrogénesis/efectos de los fármacos , Dibenzazepinas/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Receptores de Ácido Retinoico/antagonistas & inhibidores , Adipocitos/citología , Adulto , Células Madre Adultas/citología , Anciano , Cartílago Articular/citología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Condrogénesis/genética , Medio de Cultivo Libre de Suero/farmacología , Femenino , Proteínas del Grupo de Alta Movilidad/antagonistas & inhibidores , Proteínas del Grupo de Alta Movilidad/fisiología , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Osteoartritis/patología , Osteocitos/citología , ARN Interferente Pequeño/farmacología , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Ácido Retinoico/fisiología , Factor de Transcripción SOX9 , Ingeniería de Tejidos/métodos , Andamios del Tejido , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/fisiología
10.
J Tissue Eng Regen Med ; 12(6): 1402-1411, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29726103

RESUMEN

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


Asunto(s)
Ácido Ascórbico/farmacología , Diferenciación Celular/efectos de los fármacos , Condrocitos/citología , Condrogénesis , Adulto , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrogénesis/efectos de los fármacos , Condrogénesis/genética , Colágeno/metabolismo , Medios de Cultivo , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Humanos , Persona de Mediana Edad , Adulto Joven
11.
Stem Cells Transl Med ; 6(4): 1237-1248, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28186682

RESUMEN

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.


Asunto(s)
Enfermedades de los Cartílagos/terapia , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Lesiones de Menisco Tibial/terapia , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Femenino , Humanos , Técnicas In Vitro , Meniscos Tibiales/citología , Ovinos , Ingeniería de Tejidos/métodos , Andamios del Tejido , Cicatrización de Heridas/fisiología
12.
Tissue Eng ; 12(6): 1687-97, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16846363

RESUMEN

We have previously induced differentiation of embryonic stem cells (ESC) to specific phenotypes by manipulating the culture conditions, including the use of indirect co-culture. In this study, we hypothesized that co-culture with primary chondrocytes can induce human embryonic stem cells (hESC) to differentiate towards the chondrocyte lineage. Co-cultures of hESC and chondrocytes were established using well inserts, with control comprising hESC grown alone or with fibroblasts. After 28 days, after removal of the chondrocyte inserts, hESC differentiation was assessed, by morphology, immunocytochemistry, and reverse transcription polymerase chain reaction. hESC, co-cultured or grown alone, were also implanted into SCID mice on a poly-D, L-lactide scaffold, harvested 35 days later and assessed in the same way. hESC co-cultured with chondrocytes formed colonies and secreted extracellular matrix containing glycosaminoglycans (GAG). Quantitative assay showed increased synthesis of sulfated GAG in co-culture as compared with control hESC grown alone for the same period (p < 0.0001). In addition, co-cultured hESC expressed Sox 9 and collagen type II, unlike control hESC. Co-culture with fibroblasts did not induce chondrogenic differentiation. The implanted constructs with co-cultured hESC contained significantly more type II collagen (p < 0.01), type I collagen (p < 0.05), total collagen (p < 0.01), and GAG (p < 0.01) than those with hESC grown alone. Thus, we show for the first time differentiation of hESC to chondrocytes. Our results confirm the potential of the culture micro-environment to influence ESC differentiation and could provide the basis for future generation of chondrogenic cells for use in tissue repair and increase our understanding of the mechanisms that direct differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Condrocitos/citología , Condrogénesis/fisiología , Células Madre/citología , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Humanos
13.
Tissue Eng ; 12(3): 569-77, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16579690

RESUMEN

Together, the chondrocyte and its pericellular matrix have been collectively termed the chondron. Current opinion is that the pericellular matrix has both protective and signalling functions between chondrocyte and extracellular matrix. Formation of a native chondrocyte pericellular matrix or chondron structure might therefore be advantageous when tissue engineering a functional hyaline cartilage construct. The presence of chondrons has not been previously described in cartilage engineered on a scaffold. In this paper, we describe a modified immunochemical method to detect collagen VI, a key molecular marker for the pericellular matrix, and an investigation of type VI collagen distribution in engineered hyaline cartilage constructs. Cartilage constructs were engineered from adult human or bovine hyaline chondrocytes cultured on sponge or nonwoven fiber based HYAFF 11 scaffolds. Type VI collagen was detected in all constructs, but a distinctive, high-density, chondron-like distribution of collagen VI was present only in constructs exhibiting additional features of hyaline cartilage engineered using nonwoven HYAFF 11. Chondron structures were localized in areas of the extracellular matrix displaying strong collagen II and GAG staining of constructs where type II collagen composed a high percentage (over 65%) of the total collagen.


Asunto(s)
Cartílago/metabolismo , Colágeno Tipo VI/metabolismo , Ingeniería de Tejidos/métodos , Animales , Materiales Biocompatibles , Bovinos , Condrocitos/metabolismo , Colágeno Tipo II/metabolismo , Matriz Extracelular/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Ácido Hialurónico/análogos & derivados , Inmunohistoquímica , Ensayo de Materiales , Polímeros
14.
Tissue Eng ; 12(7): 1787-98, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16889509

RESUMEN

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.


Asunto(s)
Bioprótesis , Cartílago/trasplante , Condrocitos/trasplante , Ácido Hialurónico , Osteoartritis de la Rodilla/terapia , Regeneración , Ingeniería de Tejidos , Adolescente , Adulto , Cartílago/metabolismo , Condrocitos/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Tiempo
15.
Tissue Eng ; 11(1-2): 277-87, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15758586

RESUMEN

Reliable and reproducible outcome measures are essential to assess the efficacy of competing and novel tissue-engineering techniques. The aim of this study was to compare traditional histological analyses with newly developed quantitative biochemical outcome measures for the repair of articular cartilage. The production of a new anti-peptide antibody and the development and validation of a novel method for the extraction and immunoassay of type I collagen are described. The assay was used, in conjunction with existing assays for type II collagen and proteoglycans, to measure levels of the matrix components in repair tissue biopsies obtained from patients treated with the new tissue-engineering therapy Hyalograft C. Frozen sections cut from the same biopsies were stained for proteoglycans, using safranin O, and immunohistochemical analysis was used to assess type I and II collagen staining. Although there was general agreement between the extent of staining and the amounts of the three matrix components, there was a large degree of overlap in biochemical content between biopsies classified histologically on the basis of low, moderate, or abundant staining. The results demonstrate that histological grading of matrix protein abundance to classify repair cartilage as hyaline or fibrocartilagenous is often misleading. In addition, we demonstrate for the first time the ability to measure collagen cross-links in repair tissue biopsies and show that it can be used as a surrogate marker for tissue maturity. Our new range of biochemical techniques provides a standardized method to assess the quality of both engineered cartilage produced in vitro and repair tissue biopsies obtained after in vivo implantation.


Asunto(s)
Enfermedades de los Cartílagos/tratamiento farmacológico , Cartílago Articular , Condrocitos/metabolismo , Ingeniería de Tejidos/métodos , Animales , Biopsia , Enfermedades de los Cartílagos/cirugía , Bovinos , Células Cultivadas , Condrocitos/citología , Colágeno Tipo I/análisis , Colágeno Tipo I/efectos de los fármacos , Colágeno Tipo II/análisis , Colágeno Tipo II/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Secciones por Congelación , Humanos , Inmunohistoquímica , Proteoglicanos/metabolismo , Ratas , Reproducibilidad de los Resultados , Segunda Cirugía , Resultado del Tratamiento , Tripsina/farmacología
16.
Tissue Eng ; 11(9-10): 1421-8, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16259597

RESUMEN

We investigated whether, and under which conditions (i.e., cell-seeding density, medium supplements), in vitro preculture enhances in vivo development of human engineered cartilage in an ectopic nude mouse model. Monolayer-expanded adult human articular chondrocytes (AHACs) were seeded into Hyalograft C disks at 1.3 x 10(7) cells/cm3 (low density) or 7.6 x 10(7) cells/cm3 (high density). Constructs were directly implanted subcutaneously in nude mice for up to 8 weeks or precultured for 2 weeks before implantation. Preculture medium contained either transforming growth factor-beta1 (TGF-beta1, 1 ng/mL), fibroblast growth factor-2, and platelet-derived growth factor (proliferating medium) or TGF-beta1 (10 ng/mL) and insulin (differentiating medium). Both in vitro and after in vivo implantation, constructs derived by cell seeding at high versus low density and precultured in differentiating versus proliferating medium generated more cartilaginous tissues containing higher amounts of glycosaminoglycan and collagen type II and lower amounts of collagen type I, and with higher equilibrium moduli. As compared with direct implantation of freshly seeded scaffolds, preculture of AHAC-Hyalograft C constructs in differentiating medium, but not in proliferating medium, supported enhanced in vivo development of engineered cartilage. The effect of preculture was more pronounced when constructs were seeded at low density as compared with high density. This study indicates that preculture of human engineered cartilage in differentiating medium has the potential to provide grafts with higher equilibrium moduli and enhanced in vivo developmental capacity than freshly seeded scaffolds. These findings need to be validated in an orthotopic model system.


Asunto(s)
Cartílago Articular/citología , Condrocitos/citología , Condrocitos/trasplante , Ingeniería de Tejidos/métodos , Trasplante Heterotópico , Animales , Fenómenos Biofísicos , Biofisica , Cartílago Articular/fisiología , Técnicas de Cultivo de Célula , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Colágeno Tipo I/análisis , Colágeno Tipo II/análisis , Fuerza Compresiva , Medios de Cultivo/química , Medios de Cultivo/farmacología , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Glicosaminoglicanos/análisis , Histocitoquímica , Técnicas Histológicas , Humanos , Hipoglucemiantes/farmacología , Insulina/farmacología , Ratones , Ratones Desnudos , Modelos Biológicos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Factores de Tiempo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1 , Trasplante Heterólogo
17.
Cartilage ; 6(2 Suppl): 30S-5S, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27340514

RESUMEN

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.

18.
Chem Sci ; 6(11): 6106-6111, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30090225

RESUMEN

We present a new approach for the directed delivery of biomolecular payloads to individual cells with high spatial precision. This was accomplished via active sequestration of proteins, oligonucleotides or molecular dyes into coacervate microdroplets, which were then delivered to specific regions of stem cell membranes using a dynamic holographic assembler, resulting in spontaneous coacervate microdroplet-membrane fusion. The facile preparation, high sequestration efficiency and inherent membrane affinity of the microdroplets make this novel "cell paintballing" technology a highly advantageous option for spatially-directed cell functionalization, with potential applications in single cell stimulation, transfection and differentiation.

19.
Nat Commun ; 6: 7405, 2015 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-26080734

RESUMEN

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.


Asunto(s)
Cartílago Hialino , Células Madre Mesenquimatosas/efectos de los fármacos , Mioglobina/farmacología , Oxígeno/administración & dosificación , Ingeniería de Tejidos/métodos , Escherichia coli , Glicolatos/química , Humanos , Mioglobina/química
20.
Matrix Biol ; 22(3): 267-78, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12853037

RESUMEN

Cartilage oligomeric matrix protein (COMP) is a pentameric glycoprotein present in cartilage, tendon and ligament. Fragments of the molecule are present in the diseased cartilage, synovial fluid and serum of patients with knee injuries, osteoarthritis and rheumatoid arthritis. Although COMP is a substrate for several matrix metalloproteinases (MMPs), the enzymes responsible for COMP degradation in vivo have yet to be identified. In this study we utilised well-established bovine cartilage culture models to examine IL-1alpha-stimulated COMP proteolysis in the presence and absence of MMP inhibitors. COMP was released from bovine nasal cartilage, in response to IL-1alpha, at an intermediate time between proteoglycans and type II collagen, when soluble MMP levels in the culture medium were undetectable. The major fragment of COMP released following IL-1alpha-stimulation migrated with an apparent molecular mass of approximately 110 kDa (Fragment-110) and co-migrated with both the major fragment present in human arthritic synovial fluid samples and the product of COMP cleavage by purified MMP-9. However, the broad-spectrum MMP and ADAM inhibitor BB94 only partially inhibited the formation of Fragment-110 and failed to inhibit COMP release significantly. Therefore the results of these studies indicate a role for proteinases other than MMPs in the degradation of COMP in bovine cartilage. It was further demonstrated that purified COMP was cleaved by ADAMTS-4, but not ADAMTS-1 or -5, to yield a fragment which co-migrated with Fragment-110. Therefore this is the first demonstration of COMP as a substrate for ADAMTS-4, although it remains to be determined whether this enzyme plays a role in COMP degradation in vivo.


Asunto(s)
Cartílago/metabolismo , Desintegrinas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Proteínas ADAM , Proteína ADAMTS4 , Secuencias de Aminoácidos , Animales , Cartílago/efectos de los fármacos , Proteína de la Matriz Oligomérica del Cartílago , Bovinos , Técnicas de Cultivo , Desintegrinas/química , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/química , Glicoproteínas/química , Humanos , Interleucina-1/farmacología , Proteínas Matrilinas , Metaloproteinasas de la Matriz/química , Metaloendopeptidasas/metabolismo , Procolágeno N-Endopeptidasa , Líquido Sinovial/metabolismo , Trombospondinas/química , Trombospondinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA