Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Bioeng Biotechnol ; 11: 1119009, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36865027

RESUMEN

Introduction: An in vitro model that appropriately recapitulates the degenerative disc disease (DDD) microenvironment is needed to explore clinically relevant cell-based therapeutic strategies for early-stage degenerative disc disease. We developed an advanced 3D nucleus pulposus (NP) microtissues (µT) model generated with cells isolated from human degenerating NP tissue (Pfirrmann grade: 2-3), which were exposed to hypoxia, low glucose, acidity and low-grade inflammation. This model was then used to test the performance of nasal chondrocytes (NC) suspension or spheroids (NCS) after pre-conditioning with drugs known to exert anti-inflammatory or anabolic activities. Methods: NPµTs were formed by i) spheroids generated with NP cells (NPS) alone or in combination with ii) NCS or iii) NC suspension and cultured in healthy or degenerative disc disease condition. Anti-inflammatory and anabolic drugs (amiloride, celecoxib, metformin, IL-1Ra, GDF-5) were used for pre-conditioning of NC/NCS. The effects of pre-conditioning were tested in 2D, 3D, and degenerative NPµT model. Histological, biochemical, and gene expression analysis were performed to assess matrix content (glycosaminoglycans, type I and II collagen), production and release of inflammatory/catabolic factors (IL-6, IL-8, MMP-3, MMP-13) and cell viability (cleaved caspase 3). Results: The degenerative NPµT contained less glycosaminoglycans, collagens, and released higher levels of IL-8 compared to the healthy NPµT. In the degenerative NPµT, NCS performed superior compared to NC cell suspension but still showed lower viability. Among the different compounds tested, only IL-1Ra pre-conditioning inhibited the expression of inflammatory/catabolic mediators and promoted glycosaminoglycan accumulation in NC/NCS in DDD microenvironment. In degenerative NPµT model, preconditioning of NCS with IL-1Ra also provided superior anti-inflammatory/catabolic activity compared to non-preconditioned NCS. Conclusion: The degenerative NPµT model is suitable to study the responses of therapeutic cells to microenvironment mimicking early-stage degenerative disc disease. In particular, we showed that NC in spheroidal organization as compared to NC cell suspension exhibited superior regenerative performance and that IL-1Ra pre-conditioning of NCS could further improve their ability to counteract inflammation/catabolism and support new matrix production within harsh degenerative disc disease microenvironment. Studies in an orthotopic in vivo model are necessary to assess the clinical relevance of our findings in the context of IVD repair.

2.
Int J Mol Sci ; 23(13)2022 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-35805907

RESUMEN

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


Asunto(s)
Cartílago Articular , Condrocitos , Animales , Humanos , Cartílago Hialino , Hidrogeles , Ratones , Ratones Desnudos , Polietilenglicoles/farmacología , Ingeniería de Tejidos/métodos
3.
Sci Transl Med ; 13(609): eaaz4499, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34516821

RESUMEN

Osteoarthritis (OA) is the most prevalent joint disorder, causing pain and disability predominantly in the aging population but also affecting young individuals. Current treatments are limited to use of anti-inflammatory drugs to alleviate symptoms or degenerated joint replacement by a prosthetic implant at the end stage of the disease. We hypothesized that degenerative cartilage defects can be treated using nasal chondrocyte­based tissue-engineered cartilage (N-TEC). We demonstrate that N-TEC maintained cartilaginous properties when exposed in vitro to inflammatory stimuli found in osteoarthritic joints and favorably altered the inflammatory profile of cells from osteoarthritic joints. These effects were at least partially mediated by down-regulation of the WNT (wingless/integrated) signaling pathway through sFRP1 (secreted frizzled-related protein-1). We further report that N-TEC survive and engraft in vivo in ectopic mouse models reproducing a human osteochondral OA tissue environment, as well as in sheep articular cartilage defects that mimic degenerative settings. Last, we tested the safety of autologous N-TEC for the treatment of osteoarthritic cartilage defects in the knees of two patients with advanced OA (Kellgren and Lawrence grades 3 and 4) who were otherwise considered for unicondylar knee arthroplasty. No adverse reactions were recorded, and patients reported reduced pain as well as improved joint function and life quality 14 months after surgery. Together, our findings indicate that N-TEC can directly contribute to cartilage repair in osteoarthritic joints. A suitably powered clinical trial is now required to assess its efficacy in the treatment of patients with OA.


Asunto(s)
Cartílago Articular , Condrocitos , Articulación de la Rodilla , Cartílagos Nasales
4.
Acta Biomater ; 134: 240-251, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34339870

RESUMEN

Cell-based strategies for nucleus pulposus (NP) regeneration that adequately support the engraftment and functionality of therapeutic cells are still lacking. This study explores a scaffold-free approach for NP repair, which is based on spheroids derived from human nasal chondrocytes (NC), a resilient cell type with robust cartilage-regenerative capacity. We generated NC spheroids (NCS) in two types of medium (growth or chondrogenic) and analyzed their applicability for NP repair with regard to injectability, biomechanical and biochemical attributes, and integration potential in conditions simulating degenerative disc disease (DDD). NCS engineered in both media were compatible with a typical spinal needle in terms of size (lower than 600µm), shape (roundness greater than 0.8), and injectability (no changes in morphology and catabolic gene expression after passing through the needle). While growth medium ensured stable elastic modulus (E) at 5 kPa, chondrogenic medium time-dependently increased E of NCS, in correlation with gene/protein expression of collagen. Notably, DDD-mimicking conditions did not impair NCS viability nor NCS fusion with NP spheroids simulating degenerated NP in vitro. To assess the feasibility of this approach, NCS were injected into an ex vivo-cultured bovine intervertebral disc (IVD) without damage using a spinal needle. In conclusion, our data indicated that NC cultured as spheroids can be compatible with strategies for minimally invasive NP repair in terms of injectability, tuneability, biomechanical features, and resilience. Future studies will address the capacity of NCS to integrate within degenerated NP under long-term loading conditions. STATEMENT OF SIGNIFICANCE: Current regenerative strategies still do not sufficiently support the engraftment of therapeutic cells in the nucleus pulposus (NP). We present an injectable approach based on spheroids derived from nasal chondrocytes (NC), a resilient cell type with robust cartilage-regenerative capacity. NC spheroids (NCS) generated with their own matrix and demonstrated injectability, tuneability of biomechanical/biochemical attributes, and integration potential in conditions simulating degenerative disc disease. To our knowledge, this is the first study that explored an injectable spheroid-based scaffold-free approach, which showed potential to support the adhesion and viability of therapeutic cells in degenerated NP. The provided information can be of substantial interest to a wide audience, including biomaterial scientists, biomedical engineers, biologists and medical researchers.


Asunto(s)
Degeneración del Disco Intervertebral , Disco Intervertebral , Núcleo Pulposo , Animales , Bovinos , Condrocitos , Condrogénesis , Colágeno , Humanos , Degeneración del Disco Intervertebral/terapia
5.
Cartilage ; 13(2_suppl): 68S-81S, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-32959685

RESUMEN

OBJECTIVE: Cellular and molecular events occurring in cartilage regions close to injury are poorly investigated, but can possibly compromise the outcome of cell-based cartilage repair. In this study, key functional properties were assessed for cartilage biopsies collected from the central part of traumatic joint lesions (central) and from regions surrounding the defect (peripheral). These properties were then correlated with the quality of the initial cartilage biopsy and the inflammatory state of the joint. DESIGN: Cartilage samples were collected from knee joints of 42 patients with traumatic knee injuries and analyzed for cell phenotype (by reverse transcriptas-polymerase chain reaction), histological quality, cellularity, cell viability, proliferation capacity, and post-expansion chondrogenic capacity of chondrocytes (in pellet culture). Synovium was also harvested and analyzed for the expression of inflammatory cytokines. RESULTS: Cartilage quality and post-expansion chondrogenic capacity were higher in peripheral versus central samples. Differences between these 2 parameters were more pronounced in joints with high inflammatory features characterized by >100-fold difference in the mRNA levels of IL6 and IL8 in the corresponding synovium. Peripheral chondrocytes isolated from good- versus bad-quality biopsies expressed higher levels of collagen II/I and aggrecan/versican and lower levels of MMP13 and ADAMTS5. They also exhibited reduced proliferation and enhanced cartilage-forming capacity. CONCLUSIONS: Chondrocytes at the periphery of traumatic lesions better maintain properties of healthy cartilage compared to those isolated from the center, even when derived from bad-quality tissues harvested from highly inflamed joints. Future studies are necessary to investigate the change of functional properties of peripheral chondrocytes over time.


Asunto(s)
Cartílago Articular , Condrocitos , Agrecanos/metabolismo , Diferenciación Celular/genética , Condrocitos/metabolismo , Condrogénesis , Humanos
6.
Int J Mol Sci ; 18(12)2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-29186811

RESUMEN

The accumulation of senescent cells is implicated in the pathology of several age-related diseases. While the clearance of senescent cells has been suggested as a therapeutic target for patients with osteoarthritis (OA), cellular senescence of bone-resident osteoblasts (OB) remains poorly explored. Since oxidative stress is a well-known inducer of cellular senescence, we here investigated the effect of antioxidant supplementation on the isolation efficiency, expansion, differentiation potential, and transcriptomic profile of OB from osteoarthritic subchondral bone. Bone chips were harvested from sclerotic and non-sclerotic regions of the subchondral bone of human OA joints. The application of 0.1 mM ascorbic acid-2-phosphate (AA) significantly increased the number of outgrowing cells and their proliferation capacity. This enhanced proliferative capacity showed a negative correlation with the amount of senescent cells and was accompanied by decreased expression of reactive oxygen species (ROS) in cultured OB. Expanded cells continued to express differentiated OB markers independently of AA supplementation and demonstrated no changes in their capacity to osteogenically differentiate. Transcriptomic analyses revealed that apoptotic, cell cycle-proliferation, and catabolic pathways were the main pathways affected in the presence of AA during OB expansion. Supplementation with AA can thus help to expand subchondral bone OB in vitro while maintaining their special cellular characteristics. The clearance of such senescent OB could be envisioned as a potential therapeutic target for the treatment of OA.


Asunto(s)
Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Senescencia Celular , Osteoartritis/patología , Osteoblastos/efectos de los fármacos , Vitaminas/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Osteoartritis/metabolismo , Osteoblastos/metabolismo , Osteoblastos/fisiología , Especies Reactivas de Oxígeno/metabolismo , Transcriptoma
7.
Curr Opin Biotechnol ; 47: 1-6, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28551498

RESUMEN

Cells deriving from neural crest are generally acknowledged during embryonic development for their multipotency and plasticity, accounting for their capacity to generate various cell and tissue types even across germ layers. At least partial preservation of some of these properties in adulthood makes neural crest derived cells of large interest for regenerative purposes. Chondrocytes from fully mature nasal septum cartilage in adults are also derivatives of neural crest cells and were recently demonstrated to be able not only to maintain functionality across serial cloning, as surrogate self-renewal test, but also to respond and adapt to heterotopic transplantation sites. Based on these findings, cartilage grafts engineered by nasal chondrocytes were clinically used to reconstitute the nasal alar lobule and to repair articular cartilage defects. This article discusses further perspectives of potential clinical utility for nasal chondrocytes in musculoskeletal regeneration. It then highlights the need to derive deeper understanding of their biological properties in order to inform on possible therapeutic modes of action. This acquired knowledge will help to optimise manufacturing conditions to guarantee defined functional traits associated with safety and therapeutic potency of nasal chondrocytes in regenerative medicine.


Asunto(s)
Condrocitos/citología , Cresta Neural/citología , Nariz/citología , Medicina Regenerativa/métodos , Animales , Cartílago Articular/citología , Ensayos Clínicos como Asunto , Humanos
8.
Tissue Eng Part A ; 22(21-22): 1286-1295, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27633049

RESUMEN

Nasal chondrocytes (NC) were previously demonstrated to remain viable and to participate in the repair of articular cartilage defects in goats. Here, we investigated critical features of tissue-engineered grafts generated by NC in this large animal model, namely cell retention at the implantation site, architecture and integration with adjacent tissues, and effects on subchondral bone changes. In this study, isolated autologous goat NC (gNC) and goat articular chondrocytes (gAC, as control) were expanded, green fluorescent protein-labelled and seeded on a type I/III collagen membrane. After chondrogenic differentiation, tissue-engineered grafts were implanted into chondral defects (6 mm in diameter) in the stifle joint for 3 or 6 months. At the time of explantation, surrounding tissues showed no or very low (only in the infrapatellar fat pad <0.32%) migration of the grafted cells. In repair tissue, gNC formed typical structures of articular cartilage, such as flattened cells at the surface and column-like clusters in the middle layers. Semi-quantitative histological evaluation revealed efficient integration of the grafted tissues with the adjacent native cartilage and underlying subchondral bone. A significantly increased subchondral bone area, as a sign for the onset of osteoarthritis, was observed following treatment of cartilage defects with gAC-, but not with gNC-grafts. Our results reinforce the use of NC-based engineered tissue for articular cartilage repair and preliminarily indicate their potential for the treatment of early osteoarthritic defects.


Asunto(s)
Cartílago Articular , Condrocitos/metabolismo , Tabique Nasal , Regeneración , Ingeniería de Tejidos , Animales , Cartílago Articular/lesiones , Cartílago Articular/fisiología , Condrocitos/citología , Condrocitos/trasplante , Femenino , Cabras , Tabique Nasal/citología , Tabique Nasal/metabolismo
9.
Ann Transl Med ; 3(17): 254, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26605300

RESUMEN

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

10.
J Cell Mol Med ; 19(6): 1390-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25689393

RESUMEN

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


Asunto(s)
Diferenciación Celular/fisiología , Condrocitos/fisiología , Tabique Nasal/citología , Osteoblastos/fisiología , Osteogénesis/fisiología , Adulto , Anciano , Animales , Cartílago/metabolismo , Cartílago/fisiología , Diferenciación Celular/genética , Células Cultivadas , Condrocitos/metabolismo , Femenino , Expresión Génica , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones Desnudos , Persona de Mediana Edad , Osteoblastos/metabolismo , Osteogénesis/genética , Osteonectina/genética , Osteopontina/genética , Ratas Desnudas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Andamios del Tejido , Trasplante Heterólogo
11.
Sci Transl Med ; 6(251): 251ra119, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25163479

RESUMEN

In embryonic models and stem cell systems, mesenchymal cells derived from the neuroectoderm can be distinguished from mesoderm-derived cells by their Hox-negative profile--a phenotype associated with enhanced capacity of tissue regeneration. We investigated whether developmental origin and Hox negativity correlated with self-renewal and environmental plasticity also in differentiated cells from adults. Using hyaline cartilage as a model, we showed that adult human neuroectoderm-derived nasal chondrocytes (NCs) can be constitutively distinguished from mesoderm-derived articular chondrocytes (ACs) by lack of expression of specific HOX genes, including HOXC4 and HOXD8. In contrast to ACs, serially cloned NCs could be continuously reverted from differentiated to dedifferentiated states, conserving the ability to form cartilage tissue in vitro and in vivo. NCs could also be reprogrammed to stably express Hox genes typical of ACs upon implantation into goat articular cartilage defects, directly contributing to cartilage repair. Our findings identify previously unrecognized regenerative properties of HOX-negative differentiated neuroectoderm cells in adults, implying a role for NCs in the unmet clinical challenge of articular cartilage repair. An ongoing phase 1 clinical trial preliminarily indicated the safety and feasibility of autologous NC-based engineered tissues for the treatment of traumatic articular cartilage lesions.


Asunto(s)
Cartílago Articular/patología , Cresta Neural/citología , Cresta Neural/trasplante , Cicatrización de Heridas , Adulto , Animales , Cartílago Articular/citología , Proliferación Celular , Técnicas de Cocultivo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Cabras , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Articulación de la Rodilla/patología , Ratones , Persona de Mediana Edad , Plasticidad Neuronal , Proyectos Piloto , Trasplante Autólogo
12.
Tissue Eng Part C Methods ; 16(1): 11-21, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19327004

RESUMEN

We investigated whether human articular chondrocytes can be labeled efficiently and for long-term with a green fluorescent protein (GFP) lentivirus and whether the viral transduction would influence cell proliferation and tissue-forming capacity. The method was then applied to track goat articular chondrocytes after autologous implantation in cartilage defects. Expression of GFP in transduced chondrocytes was detected cytofluorimetrically and immunohistochemically. Chondrogenic capacity of chondrocytes was assessed by Safranin-O staining, immunostaining for type II collagen, and glycosaminoglycan content. Human articular chondrocytes were efficiently transduced with GFP lentivirus (73.4 +/- 0.5% at passage 1) and maintained the expression of GFP up to 22 weeks of in vitro culture after transduction. Upon implantation in nude mice, 12 weeks after transduction, the percentage of labeled cells (73.6 +/- 3.3%) was similar to the initial one. Importantly, viral transduction of chondrocytes did not affect the cell proliferation rate, chondrogenic differentiation, or tissue-forming capacity, either in vitro or in vivo. Goat articular chondrocytes were also efficiently transduced with GFP lentivirus (78.3 +/- 3.2%) and maintained the expression of GFP in the reparative tissue after orthotopic implantation. This study demonstrates the feasibility of efficient and relatively long-term labeling of human chondrocytes for co-culture on integration studies, and indicates the potential of this stable labeling technique for tracking animal chondrocytes for in cartilage repair studies.


Asunto(s)
Condrocitos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Lentivirus/genética , Animales , Diferenciación Celular , Proliferación Celular , Condrocitos/citología , Técnicas de Cocultivo , Colágeno Tipo II/metabolismo , Glicosaminoglicanos/metabolismo , Cabras , Humanos , Inmunohistoquímica/métodos , Ratones , Ratones Desnudos , Fenazinas/farmacología , Reproducibilidad de los Resultados
13.
Swiss Med Wkly ; 139(41-42): 602-9, 2009 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-19918699

RESUMEN

The in vitro engineering of functionally developed biological cartilage substitutes, based on cells and appropriate structural and soluble factors, is an attractive concept for the clinical treatment of cartilage injuries and degeneration. The field of cartilage tissue engineering has developed strongly in the last few years, bringing together the scientific, clinical and commercial interests of highly interdisciplinary communities. However, engineered grafts are still far from being the standard of care for cartilage repair. In this review we present some of the issues challenging the reproducible engineering of functional cartilage templates starting from human cells. We then discuss the need to identify the mode of action of cartilage tissue engineering approaches, which in turn is expected to define potency markers and quality controls for grafts capable of inducing durable cartilage regeneration. Finally, we propose the use of engineered cartilage tissues not only as implants to be implemented in the clinic, but also as models to understand mechanisms and processes related to cartilage development and repair. The knowledge generated using these models will be instrumental in moving to the next generation of cartilage repair approaches, namely those inducing regeneration in situ, based on the recruitment of resident cells.


Asunto(s)
Cartílago/trasplante , Ingeniería de Tejidos/métodos , Andamios del Tejido , Humanos
14.
J Cell Physiol ; 219(1): 219-26, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19107842

RESUMEN

A current challenge in mesenchymal stem cell (MSC)-based cartilage repair is to solve donor and tissue-dependent variability of MSC cultures and to prevent chondrogenic cells from terminal differentiation like in the growth plate. The aim of this study was to select the best source for MSC which could promise stable cartilage formation in the absence of hypertrophy and ectopic in vivo mineralization. We hypothesized that MSC from synovium are superior to bone marrow- and adipose tissue-derived MSC since they are derived from a joint tissue. MSC were characterized by flow cytometry. MSC pellets were cultured under chondrogenic conditions and differentiation was evaluated by histology, gene expression analysis, and determination of alkaline phosphatase activity (ALP). After chondrogenic induction, pellets were transplanted subcutaneously into SCID mice. MSC from bone marrow, adipose tissue, and synovium revealed similar COL2A1/COL10A1 mRNA levels after chondrogenic induction and were positive for collagen-type-X. Bone marrow-derived and adipose tissue-derived MSC showed significantly higher ALP activity than MSC from synovium. Low ALP-activity before transplantation of pellets correlated with marginal calcification of explants. Surprisingly, non-mineralizing transplants specifically lost their collagen-type II, but not collagen-type I deposition in vivo, or were fully degraded. In conclusion, the lower donor-dependent ALP activation and reduced mineralization of synovium-derived heterotopic transplants did not lead to stable ectopic cartilage as known from articular chondrocytes, but correlated with fibrous dedifferentiation or complete degeneration of MSC pellets. This emphasizes that beside appropriate induction of differentiation, locking of MSC in the desired differentiation state is a major challenge for MSC-based repair strategies.


Asunto(s)
Calcificación Fisiológica , Diferenciación Celular/fisiología , Condrogénesis/fisiología , Células Madre Mesenquimatosas/fisiología , Adipocitos/citología , Adipocitos/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Fosfatasa Alcalina/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Trasplante de Células , Células Cultivadas , Condrocitos/citología , Condrocitos/fisiología , Colágeno Tipo II/metabolismo , Femenino , Humanos , Masculino , Metaloproteinasas de la Matriz/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Ratones SCID , Persona de Mediana Edad , Membrana Sinovial/citología , Adulto Joven
15.
Injury ; 39 Suppl 1: S58-65, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18313473

RESUMEN

The application of autologous chondrocytes in cartilage repair procedures is associated with several disadvantages, including injury of healthy cartilage in a preceding surgery frequently resulting in formation of inferior fibrocartilage at defect sites. In order to improve the quality of regeneration, adult mesenchymal stem cells (MSC) are regarded as a promising alternative. The great challenge, when considering MSC for articular cartilage repair, is to generate cells with features of stable chondrocytes which are resistant to hypertrophy and terminal differentiation, as found in hyaline articular cartilage. Common in vitro protocols for chondrogenic differentiation of MSC successfully induce expression of multiple cartilage-specific molecules, including collagen type II and aggrecan, and result in a chondrocyte-like phenotype. However, in vitro chondrogenesis of MSC additionally promotes induction of fibrocartilage-like features such as expression of collagen type I, and hypertrophy, as demonstrated by up-regulation of collagen type X, MMP13 and ALP-activity. As a consequence, differentiated MSC pellets undergo mineralisation and vascularisation after ectopic transplantation in a process similar to endochondral ossification. This review discusses the complexity and entailed challenges when considering MSC from various sources for clinical application and the necessity to optimise chondrogenesis by repressing hypertrophy to obtain functional and suitable cells for cartilage repair.


Asunto(s)
Cartílago Articular/metabolismo , Condrocitos/metabolismo , Condrogénesis/fisiología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Animales , Cartílago Articular/citología , Diferenciación Celular/fisiología , Condrocitos/citología , Condrogénesis/genética , Humanos , Células Madre Mesenquimatosas/citología , Conejos , Ingeniería de Tejidos/métodos
16.
Arthritis Rheum ; 58(2): 467-74, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18240244

RESUMEN

OBJECTIVE: Monolayer expansion of human articular chondrocytes (HACs) is known to result in progressive dedifferentiation of the chondrocytes and loss of their stable cartilage formation capacity in vivo. For an optimal outcome of chondrocyte-based repair strategies, HACs capable of ectopic cartilage formation may be required. This study was undertaken to identify secreted candidate molecules, in supernatants of cultured HACs, that could serve as predictors of the ectopic cartilage formation capacity of cells. METHODS: Standardized medium supernatants (n = 5 knee cartilage samples) of freshly isolated HACs (PD0) and of HACs expanded for 2 or 6 population doublings (PD2 and PD6, respectively) were screened by a multiplexed immunoassay for 15 distinct interleukins, 8 matrix metalloproteinases (MMPs), and 11 miscellaneous soluble factors. Cartilage differentiation markers such as cartilage oligomeric matrix protein and YKL-40 were determined by enzyme-linked immunosorbent assay. HACs from each culture were subcutaneously transplanted into SCID mice, and the capacity of the chondrocytes to form stable cartilage was examined histologically 4 weeks later. RESULTS: Whereas freshly isolated (PD0) HACs generated stable ectopic cartilage that was positive for type II collagen, none of the cell transplants at PD6 formed cartilaginous matrix. Loss of the ectopic cartilage formation capacity between PD0 and PD6 correlated with a drop in the secretion of MMP-3 to <10% of initial levels, whereas changes in the other investigated molecules were not predictive. Chondrocytes with MMP-3 levels of >or=20% of initial levels synthesized cartilaginous matrix, whereas those with low MMP-3 levels (<10% of initial levels) at PD2 failed to regenerate ectopic cartilage. CONCLUSION: Loss of the capacity for stable ectopic cartilage formation in the course of HAC dedifferentiation can be predicted by determining the relative levels of MMP-3, demonstrating that standardized culture supernatants can be used for quality control of chondrocytes dedicated for cell therapeutic approaches.


Asunto(s)
Cartílago Articular , Condrocitos/enzimología , Condrocitos/trasplante , Coristoma/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Trasplante de Células/normas , Células Cultivadas , Niño , Condrocitos/citología , Coristoma/patología , Humanos , Ratones , Ratones SCID , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Control de Calidad
17.
Matrix Biol ; 26(1): 30-41, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17074475

RESUMEN

Cartilage acidic protein 1 (CRTAC1), a novel human marker which allowed discrimination of human chondrocytes from osteoblasts and mesenchymal stem cells in culture was so far studied only on the RNA-level. We here describe its genomic organisation and detect a new brain expressed (CRTAC1-B) isoform resulting from alternate last exon usage which is highly conserved in vertebrates. In humans, we identify an exon sharing process with the neighbouring tail-to-tail orientated gene leading to CRTAC1-A. This isoform is produced by cultured human chondrocytes, localized in the extracellular matrix of articular cartilage and its secretion can be stimulated by BMP4. Of five putative O-glycosylation motifs in the last exon of CRTAC1-A, the most C-terminal one is modified according to exposure of serial C-terminal deletion mutants to the O-glycosylation inhibitor Benzyl-alpha-GalNAc. Both isoforms contain four FG-GAP repeat domains and an RGD integrin binding motif, suggesting cell-cell or cell-matrix interaction potential. In summary, CRTAC1 acquired an alternate last exon from the tail-to-tail oriented neighbouring gene in humans resulting in the glycosylated isoform CRTAC1-A which represents a new extracellular matrix molecule of articular cartilage.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Acetilgalactosamina/análogos & derivados , Acetilgalactosamina/farmacología , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Compuestos de Bencilo/farmacología , Sitios de Unión , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/farmacología , Encéfalo/metabolismo , Proteínas de Unión al Calcio/genética , Cartílago Articular/citología , Niño , Condrocitos/efectos de los fármacos , Colágeno Tipo II/metabolismo , Inhibidores Enzimáticos/farmacología , Exones , Proteínas de la Matriz Extracelular/genética , Glicosilación/efectos de los fármacos , Humanos , Pulmón/metabolismo , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Homología de Secuencia de Aminoácido , Treonina/metabolismo , Vertebrados/genética
18.
Arthritis Rheum ; 54(10): 3254-66, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17009260

RESUMEN

OBJECTIVE: Functional suitability and phenotypic stability of ectopic transplants are crucial factors in the clinical application of mesenchymal stem cells (MSCs) for articular cartilage repair, and might require a stringent control of chondrogenic differentiation. This study evaluated whether human bone marrow-derived MSCs adopt natural differentiation stages during induction of chondrogenesis in vitro, and whether they can form ectopic stable cartilage that is resistant to vascular invasion and calcification in vivo. METHODS: During in vitro chondrogenesis of MSCs, the expression of 44 cartilage-, stem cell-, and bone-related genes and the deposition of aggrecan and types II and X collagen were determined. Similarly treated, expanded articular chondrocytes served as controls. MSC pellets were allowed to differentiate in chondrogenic medium for 3-7 weeks, after which the chondrocytes were implanted subcutaneously into SCID mice; after 4 weeks in vivo, samples were evaluated by histology. RESULTS: The 3-stage chondrogenic differentiation cascade initiated in MSCs was primarily characterized by sequential up-regulation of common cartilage genes. Premature induction of hypertrophy-related molecules (type X collagen and matrix metalloproteinase 13) occurred before production of type II collagen and was followed by up-regulation of alkaline phosphatase activity. In contrast, hypertrophy-associated genes were not induced in chondrocyte controls. Whereas control chondrocyte pellets resisted calcification and vascular invasion in vivo, most MSC pellets mineralized, in spite of persisting proteoglycan and type II collagen content. CONCLUSION: An unnatural pathway of differentiation to chondrocyte-like cells was induced in MSCs by common in vitro protocols. MSC pellets transplanted to ectopic sites in SCID mice underwent alterations related to endochondral ossification rather than adopting a stable chondrogenic phenotype. Further studies are needed to evaluate whether a more stringent control of MSC differentiation to chondrocytes can be achieved during cartilage repair in a natural joint environment.


Asunto(s)
Calcinosis/patología , Cartílago Articular/irrigación sanguínea , Cartílago Articular/patología , Condrocitos/patología , Coristoma/patología , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/patología , Adulto , Anciano , Agrecanos/genética , Agrecanos/metabolismo , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Calcinosis/genética , Calcinosis/metabolismo , Cartílago Articular/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Condrocitos/metabolismo , Condrocitos/trasplante , Coristoma/genética , Coristoma/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Colágeno Tipo X/genética , Colágeno Tipo X/metabolismo , Femenino , Humanos , Hipertrofia/genética , Hipertrofia/metabolismo , Hipertrofia/fisiopatología , Masculino , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones SCID , Persona de Mediana Edad , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA