Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cytotherapy ; 25(3): 286-297, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36599772

RESUMEN

BACKGROUND AIMS: Cell therapies have the potential to improve reconstructive procedures for congenital craniofacial cartilage anomalies such as microtia. Adipose-derived stem cells (ADSCs) and auricular cartilage stem/progenitor cells (CSPCs) are promising candidates for cartilage reconstruction, but their successful use in the clinic will require the development of xeno-free expansion and differentiation protocols that can maximize their capacity for chondrogenesis. METHODS: We assessed the behavior of human ADSCs and CSPCs grown either in qualified fetal bovine serum (FBS) or human platelet lysate (hPL), a xeno-free alternative, in conventional monolayer and 3-dimensional spheroid cultures. RESULTS: We show that CSPCs and ADSCs display greater proliferation rate in hPL than FBS and express typical mesenchymal stromal cell surface antigens in both media. When expanded in hPL, both cell types, particularly CSPCs, maintain a spindle-like morphology and lower surface area over more passages than in FBS. Both media supplements support chondrogenic differentiation of CSPCs and ADSCs grown either as monolayers or spheroids. However, chondrogenesis appears less ordered in hPL than FBS, with reduced co-localization of aggrecan and collagen type II in spheroids. CONCLUSIONS: hPL may be beneficial for the expansion of cells with chondrogenic potential and maintaining stemness, but not for their chondrogenic differentiation for tissue engineering or disease modeling.


Asunto(s)
Adipocitos , Condrogénesis , Humanos , Niño , Diferenciación Celular , Células Cultivadas , Proliferación Celular , Plaquetas
2.
Brain Commun ; 6(4): fcae225, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38983619

RESUMEN

Members of the phosphodiesterase 4 (PDE4) enzyme family regulate the availability of the secondary messenger cyclic adenosine monophosphate (cAMP) and, by doing so, control cellular processes in health and disease. In particular, PDE4D has been associated with Alzheimer's disease and the intellectual disability seen in fragile X syndrome. Furthermore, single point mutations in critical PDE4D regions cause acrodysostosis type 2(ACRDYS2, also referred to as inactivating PTH/PTHrP signalling disorder 5 or iPPSD5), where intellectual disability is seen in ∼90% of patients alongside the skeletal dysmorphologies that are characteristic of acrodysostosis type 1 (ACRDYS1/iPPSD4) and ACRDYS2. Two contrasting mechanisms have been proposed to explain how mutations in PDE4D cause iPPSD5. The first mechanism, the 'over-activation hypothesis', suggests that cAMP/PKA (cyclic adenosine monophosphate/protein kinase A) signalling is reduced by the overactivity of mutant PDE4D, whilst the second, the 'over-compensation hypothesis' suggests that mutations reduce PDE4D activity. That reduction in activity is proposed to cause an increase in cellular cAMP, triggering the overexpression of other PDE isoforms. The resulting over-compensation then reduces cellular cAMP and the levels of cAMP/PKA signalling. However, neither of these proposed mechanisms accounts for the fine control of PDE activation and localization, which are likely to play a role in the development of iPPSD5. This review will draw together our understanding of the role of PDE4D in iPPSD5 and present a novel perspective on possible mechanisms of disease.

3.
PLoS One ; 18(11): e0294761, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37992123

RESUMEN

Reconstruction of the outer ear currently requires harvesting of cartilage from the posterior of the auricle or ribs leading to pain and donor site morbidity. An alternative source for auricular reconstruction is in vitro tissue engineered cartilage using stem/progenitor cells. Several candidate cell-types have been studied with tissue-specific auricular cartilage progenitor cells (AuCPC) of particular interest. Whilst chondrogenic differentiation of competent stem cells using growth factor TGFß1 produces cartilage this tissue is frequently fibrocartilaginous and lacks the morphological features of hyaline cartilage. Recent work has shown that growth factor BMP9 is a potent chondrogenic and morphogenetic factor for articular cartilage progenitor cells, and we hypothesised that this property extends to cartilage-derived progenitors from other tissues. In this study we show monoclonal populations of AuCPCs from immature and mature bovine cartilage cultured with BMP9 produced cartilage pellets have 3-5-fold greater surface area in sections than those grown with TGFß1. Increased volumetric growth using BMP9 was due to greater sGAG deposition in immature pellets and significantly greater collagen accumulation in both immature and mature progenitor pellets. Polarised light microscopy and immunohistochemical analyses revealed that the organisation of collagen fibrils within pellets is an important factor in the growth of pellets. Additionally, chondrocytes in BMP9 stimulated cell pellets had larger lacunae and were more evenly dispersed throughout the extracellular matrix. Interestingly, BMP9 tended to normalise the response of immature AuCPC monoclonal cell lines to differentiation cues whereas cells exhibited more variation under TGFß1. In conclusion, BMP9 appears to be a potent inducer of chondrogenesis and volumetric growth for AuCPCs a property that can be exploited for tissue engineering strategies for reconstructive surgery though with the caveat of negligible elastin production following 21-day treatment with either growth factor.


Asunto(s)
Cartílago Articular , Cartílago Auricular , Animales , Bovinos , Colágeno Tipo II/metabolismo , Condrogénesis/fisiología , Condrocitos/metabolismo , Diferenciación Celular/fisiología , Cartílago Articular/metabolismo , Colágeno/metabolismo , Células Cultivadas
4.
Stem Cells Transl Med ; 9(12): 1651-1666, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32639692

RESUMEN

While human adipose-derived stem cells (hADSCs) are known to possess osteogenic differentiation potential, the bone tissues formed are generally considered rudimentary and immature compared with those made by bone-derived precursor cells such as human bone marrow-derived mesenchymal stem cells (hBMSCs) and less commonly studied human calvarium osteoprogenitor cells (hOPs). Traditional differentiation protocols have tended to focus on osteoinduction of hADSCs through the addition of osteogenic differentiation media or use of stimulatory bioactive scaffolds which have not resulted in mature bone formation. Here, we tested the hypothesis that by reproducing the physical as well as biochemical bone microenvironment through the use of three-dimensional (3D) culture and vascularization we could enhance osteogenic maturation in hADSCs. In addition to biomolecular characterization, we performed structural analysis through extracellular collagen alignment and mineral density in our bone tissue engineered samples to evaluate osteogenic maturation. We further compared bone formed by hADSCs, hBMSCs, and hOPs against mature human pediatric calvarial bone, yet not extensively investigated. Although bone generated by all three cell types was still less mature than native pediatric bone, a fibrin-based 3D microenvironment together with vascularization boosted osteogenic maturation of hADSC making it similar to that of bone-derived osteoprogenitors. This demonstrates the important role of vascularization and 3D culture in driving osteogenic maturation of cells easily available but constitutively less committed to this lineage and suggests a crucial avenue for recreating the bone microenvironment for tissue engineering of mature craniofacial bone tissues from pediatric hADSCs, as well as hBMSCs and hOPs.


Asunto(s)
Tejido Adiposo/metabolismo , Osteogénesis/fisiología , Células Madre/metabolismo , Ingeniería de Tejidos/métodos , Humanos , Andamios del Tejido
5.
Stem Cells Dev ; 29(14): 882-894, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32364057

RESUMEN

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


Asunto(s)
Cartílago Articular/citología , Condrogénesis , Factor 2 de Diferenciación de Crecimiento/metabolismo , Células Madre/citología , Animales , Bovinos , Células Cultivadas , Colágeno/metabolismo , Regulación de la Expresión Génica , Factor 2 de Diferenciación de Crecimiento/genética , Hidroxiprolina/metabolismo
6.
Tissue Eng Part A ; 24(9-10): 849-859, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29073831

RESUMEN

INTRODUCTION: Mesenchymal stem cells (MSCs) are considered a promising cell source for cartilage repair strategies due to their chondrogenic differentiation potential. However, their in vitro tendency to progress toward hypertrophy limits their clinical use. This unfavorable result may be due to the fact that MSCs used in tissue engineering approaches are all at the same developmental stage, and have lost crucial spatial and temporal signaling cues. In this study, we sought to investigate the effect of a spatial parathyroid hormone-related protein (PTHrP) signaling gradient on the chondrogenic differentiation of MSCs and progression to hypertrophy. METHODS: Human bone marrow-derived MSCs were transduced with adenoviral vectors overexpressing PTHrP and seeded into fibrin-poly(ester-urethane) scaffolds. To investigate the effect of a spatial PTHrP signaling gradient, scaffolds were seeded with PTHrP-overexpressing MSCs positioned on top of the scaffold, with untransduced MSCs seeded evenly within. Scaffolds were cultured with or without 2 ng/mL transforming growth factor (TGF)-ß1 for 28 days. RESULTS: PTHrP overexpression increased glycosaminoglycan (GAG) production by MSCs irrespective of TGF-ß1 treatment, and exerted differential effects on chondrogenic and hypertrophic gene expression when MSCs were cultured in the presence of a PTHrP signaling gradient. Furthermore, PTHrP-overexpressing MSCs were associated with an increase of endogenous TGF-ß1 production and reduced total MMP-13 secretion compared to controls. CONCLUSION: The presence of a spatial PTHrP signaling gradient may support chondrogenic differentiation of MSCs and promote the formation of a more stable cartilage phenotype in tissue engineering applications.


Asunto(s)
Condrogénesis/fisiología , Células Madre Mesenquimatosas/citología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Adenoviridae/genética , Fosfatasa Alcalina/metabolismo , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos/genética , Humanos , Hipertrofia/metabolismo , Ingeniería de Tejidos/métodos
7.
J Tissue Eng Regen Med ; 11(9): 2663-2666, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-27445107

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) is widely used in an active recombinant form to stimulate the chondrogenic differentiation of mesenchymal stem cells (MSCs). Recently, it has been shown that the application of multiaxial load, that mimics the loading within diarthrodial joints, to MSCs seeded in to fibrin-poly(ester-urethane) scaffolds leads to the endogenous production and secretion of TGF-ß1 by the mechanically stimulated cells, which in turn drives the chondrogenic differentiation of the cells within the scaffold. The work presented in this short communication provides further evidence that the application of joint mimicking multiaxial load induces the secretion of TGF-ß1 by mechanically stimulated MSCs. The results of this work also show that joint-like multiaxial mechanical load activates latent TGF-ß1 in response to loading in the presence or absence of cells; this activation was not seen in non-loaded control scaffolds. Despite the application of mechanical load to scaffolds with different distributions/numbers of cells no significant differences were seen in the percentage of active TGF-ß1 quantified in the culture medium of scaffolds from different groups. The similar level of activation in scaffolds containing different numbers of cells, cells at different stages of differentiation or with different distributions of cells suggests that this activation results from the mechanical forces applied to the culture system rather than differences in cellular behaviour. These results are relevant when considering rehabilitation protocols after cell therapy or microfracture, for articular cartilage repair, where increased TGF-ß1 activation in response to joint mobilization may improve the quality of developing cartilaginous repair material. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Fibrina/química , Células Madre Mesenquimatosas/metabolismo , Poliésteres/química , Poliuretanos/química , Estrés Mecánico , Andamios del Tejido/química , Factor de Crecimiento Transformador beta1/metabolismo , Adolescente , Adulto , Femenino , Humanos , Articulaciones , Masculino , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad
8.
J Tissue Eng Regen Med ; 11(10): 2912-2921, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-27406210

RESUMEN

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


Asunto(s)
Condrogénesis/efectos de los fármacos , Fibrina/farmacología , Células Madre Mesenquimatosas/citología , Poliésteres/farmacología , Poliuretanos/farmacología , Andamios del Tejido/química , Adolescente , Anciano , ADN/metabolismo , Femenino , Colorantes Fluorescentes/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Coloración y Etiquetado , Adulto Joven
9.
Methods Mol Biol ; 1340: 41-52, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26445829

RESUMEN

Mesenchymal stem cells are found in a number of tissues and have the potential to differentiate into a range of mesenchymal lineages. This ready availability and multipotent character means that mesenchymal stem cells have become a focus for the field of tissue engineering, particularly for the repair of bone and cartilage. This chapter describes the isolation of mesenchymal stem cells from human bone marrow tissue, as well as expansion of the cells and characterisation of their multipotency.


Asunto(s)
Células de la Médula Ósea/fisiología , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Separación Celular/métodos , Células Madre Mesenquimatosas/fisiología , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Adipogénesis , Biomarcadores/metabolismo , Células de la Médula Ósea/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Condrogénesis , Humanos , Cinética , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Fenotipo , Coloración y Etiquetado
10.
PLoS One ; 10(8): e0136229, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26292283

RESUMEN

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


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Cartílago Articular/citología , Células Madre/citología , Estrés Mecánico , Regulación hacia Arriba , Adenoviridae/genética , Células Cultivadas , Condrogénesis , Colágeno Tipo II/metabolismo , Fibrina/química , Regulación de la Expresión Génica , Vectores Genéticos/genética , Glicosaminoglicanos/metabolismo , Humanos , Células Madre/metabolismo , Andamios del Tejido/química , Transducción Genética , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA