Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomaterials ; 276: 120996, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34280823

RESUMEN

One of the major challenges within the emerging field of injectable stem cell therapies for articular cartilage (AC) repair is the retention of sufficient viable cell numbers at the site of injury. Even when delivered via intra-articular injection, the number of stem cells retained at the target is often low and declines rapidly over time. To address this challenge, an artificial plasma membrane binding nanocomplex was rationally designed to provide human mesenchymal stem cells (hMSCs) with increased adhesion to articular cartilage tissue. The nanocomplex comprises the extracellular matrix (ECM) binding peptide of a placenta growth factor-2 (PlGF-2) fused to a supercharged green fluorescent protein (scGFP), which was electrostatically conjugated to anionic polymer surfactant chains to yield [S-]scGFP_PlGF2. The [S-]scGFP_PlGF2 nanocomplex spontaneously inserts into the plasma membrane of hMSCs, is not cytotoxic, and does not inhibit differentiation. The nanocomplex-modified hMSCs showed a significant increase in affinity for immobilised collagen II, a key ECM protein of cartilage, in both static and dynamic cell adhesion assays. Moreover, the cells adhered strongly to bovine ex vivo articular cartilage explants resulting in high cell numbers. These findings suggest that the re-engineering of hMSC membranes with [S-]scGFP_PlGF2 could improve the efficacy of injectable stem cell-based therapies for the treatment of damaged articular cartilage.


Asunto(s)
Cartílago Articular , Células Madre Mesenquimatosas , Animales , Proteínas Portadoras , Bovinos , Adhesión Celular , Diferenciación Celular , Condrogénesis , Matriz Extracelular , Humanos , Membranas Artificiales , Polímeros , Células Madre , Tensoactivos
2.
Artículo en Inglés | MEDLINE | ID: mdl-32266231

RESUMEN

Smart biomaterials with an inherent capacity to elicit specific behaviors in lieu of biological prompts would be advantageous for regenerative medicine applications. In this work, we employ an electrospinning technique to model the in vivo nanofibrous extracellular matrix (ECM) of cartilage using a chondroinductive cellulose and silk polymer blend (75:25 ratio). This natural polymer composite is directly electrospun for the first time, into nanofibers without post-spun treatment, using a trifluoroacetic acid and acetic acid cosolvent system. Biocompatibility of the composite nanofibres with human mesenchymal stem cells (hMSCs) is demonstrated and its inherent capacity to direct chondrogenic stem cell differentiation, in the absence of stimulating growth factors, is confirmed. This chondrogenic stimulation could be countered biochemically using fibroblast growth factor-2, a growth factor used to enhance the proliferation of hMSCs. Furthermore, the potential mechanisms driving this chondroinduction at the cell-biomaterial interface is investigated. Composite substrates are fabricated as two-dimensional film surfaces and cultured with hMSCs in the presence of chemicals that interfere with their biochemical and mechanical signaling pathways. Preventing substrate surface elasticity transmission resulted in a significant downregulation of chondrogenic gene expression. Interference with the classical chondrogenic Smad2/3 phosphorylation pathway did not impact chondrogenesis. The results highlight the importance of substrate mechanical elasticity on hMSCs chondroinduction and its independence to known chondrogenic biochemical pathways. The newly fabricated scaffolds provide the foundation for designing a robust, self-inductive, and cost-effective biomimetic biomaterial for cartilage tissue engineering.

3.
Cell Prolif ; 52(6): e12653, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31489992

RESUMEN

OBJECTIVES: Bioreactor-based production systems have the potential to overcome limitations associated with conventional tissue engineering manufacturing methods, facilitating regulatory compliant and cost-effective production of engineered grafts for widespread clinical use. In this work, we established a bioreactor-based manufacturing system for the production of cartilage grafts. MATERIALS & METHODS: All bioprocesses, from cartilage biopsy digestion through the generation of engineered grafts, were performed in our bioreactor-based manufacturing system. All bioreactor technologies and cartilage tissue engineering bioprocesses were transferred to an independent GMP facility, where engineered grafts were manufactured for two large animal studies. RESULTS: The results of these studies demonstrate the safety and feasibility of the bioreactor-based manufacturing approach. Moreover, grafts produced in the manufacturing system were first shown to accelerate the repair of acute osteochondral defects, compared to cell-free scaffold implants. We then demonstrated that grafts produced in the system also facilitated faster repair in a more clinically relevant chronic defect model. Our data also suggested that bioreactor-manufactured grafts may result in a more robust repair in the longer term. CONCLUSION: By demonstrating the safety and efficacy of bioreactor-generated grafts in two large animal models, this work represents a pivotal step towards implementing the bioreactor-based manufacturing system for the production of human cartilage grafts for clinical applications. Read the Editorial for this article on doi:10.1111/cpr.12625.


Asunto(s)
Reactores Biológicos , Condrocitos/citología , Ingeniería de Tejidos , Andamios del Tejido , Enfermedad Aguda , Animales , Cartílago Articular/patología , Enfermedad Crónica , Femenino , Modelos Animales , Ovinos , Ingeniería de Tejidos/métodos
4.
Mater Sci Eng C Mater Biol Appl ; 93: 254-264, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30274057

RESUMEN

The aim of this study was to investigate the synthesis and in vitro characterization of thermoset biodegradable poly (diol-co-tricarballylate) (PDT) elastomeric polymers for the purpose of their use in implantable drug delivery and tissue engineering applications. The synthesis was based on thermal crosslinking technique via a polycondensation reaction of tricarballylic acid with aliphatic diols of varying chain lengths (C6-C12). PDT prepolymers were synthesized at 140 °C for 20 min. After purification, the prepolymers were molded and kept at 120 °C for 18 h under vacuum to complete the crosslinking process. PDT prepolymers were characterized by DSC, FT-IR, 1H NMR and GPC. The PDT elastomers were also subjected to thermal and structural analysis, as well as sol content, mechanical testing, in vitro degradation and cytocompatibility studies. The mechanical properties and sol content were found to be dependent on synthesis conditions and can be controlled by manipulating the crosslinking density and number of methylene groups in the chain of precursor aliphatic diol. The family of thermally crosslinked PDT biodegradable polyesters were successfully prepared and characterized; besides they have promising use in drug delivery and other biomedical tissue engineering applications.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Sistemas de Liberación de Medicamentos/métodos , Elastómeros/química , Ensayo de Materiales , Ingeniería de Tejidos , Animales , Línea Celular Tumoral , Ratones
5.
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
6.
Polymers (Basel) ; 10(4)2018 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-30966490

RESUMEN

Reactive electrospinning is capable of efficiently producing in situ crosslinked scaffolds resembling the natural extracellular matrix with tunable characteristics. In this study, we aimed to synthesize, characterize, and investigate the in vitro cytocompatibility of electrospun fibers of acrylated poly(1,10-decanediol-co-tricarballylate) copolymer prepared utilizing the photoreactive electrospinning process with ultraviolet radiation for crosslinking, to be used for cardiac tissue engineering applications. Chemical, thermal, and morphological characterization confirmed the successful synthesis of the polymer used for production of the electrospun fibrous scaffolds with more than 70% porosity. Mechanical testing confirmed the elastomeric nature of the fibers required to withstand cardiac contraction and relaxation. The cell viability assay showed no significant cytotoxicity of the fibers on cultured cardiomyoblasts and the cell-scaffolds interaction study showed a significant increase in cell attachment and growth on the electrospun fibers compared to the reference. This data suggests that the newly synthesized fibrous scaffold constitutes a promising candidate for cardiac tissue engineering applications.

7.
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
8.
Redox Biol ; 12: 483-490, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28334683

RESUMEN

OBJECTIVE: Obesity-associated impaired fat accumulation in the visceral adipose tissue can lead to ectopic fat deposition and increased risk of insulin resistance and type 2 diabetes mellitus (T2DM). This study investigated whether impaired adipogenesis of omental (OM) adipose tissues and elevated 4-hydroxynonenal (4-HNE) accumulation contribute to this process, and if combined metformin and insulin treatment in T2DM patients could rescue this phenotype. METHODS: OM adipose tissues were obtained from forty clinically well characterized obese individuals during weight reduction surgery. Levels of 4-HNE protein adducts, adipocyte size and number of macrophages were determined within these tissues by immunohistochemistry. Adipogenic capacity and gene expression profiles were assessed in preadipocytes derived from these tissues in relation to insulin resistance and in response to 4-HNE, metformin or combined metformin and insulin treatment. RESULTS: Preadipocytes isolated from insulin resistant (IR) and T2DM individuals exhibited lower adipogenesis, marked by upregulation of anti-adipogenic genes, compared to preadipocytes derived from insulin sensitive (IS) individuals. Impaired adipogenesis was also associated with increased 4-HNE levels, smaller adipocytes and greater macrophage presence in the adipose tissues. Within the T2DM group, preadipocytes from combined metformin and insulin treated subset showed better in vitro adipogenesis compared to metformin alone, which was associated with less presence of macrophages and 4-HNE in the adipose tissues. Treatment of preadipocytes in vitro with 4-HNE reduced their adipogenesis and increased proliferation, even in the presence of metformin, which was partially rescued by the presence of insulin. CONCLUSION: This study reveals involvement of 4-HNE in the impaired OM adipogenesis-associated with insulin resistance and T2DM and provides a proof of concept that this impairment can be reversed by the synergistic action of insulin and metformin. Further studies are needed to evaluate involvement of 4-HNE in metabolically impaired abdominal adipogenesis and to confirm benefits of combined metformin-insulin therapy in T2DM patients.


Asunto(s)
Adipogénesis/efectos de los fármacos , Aldehídos/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Insulina/farmacología , Metformina/farmacología , Obesidad/metabolismo , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adulto , Cirugía Bariátrica , Células Cultivadas , Diabetes Mellitus Tipo 2/cirugía , Quimioterapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Obesidad/cirugía
9.
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
10.
Free Radic Biol Med ; 104: 129-137, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28088621

RESUMEN

OBJECTIVE: Increased adipose production of 4-hydroxynonenal (4-HNE), a bioreactive aldehyde, directly correlates with obesity and insulin resistance. The aim of this study was to elucidate the impact of 4-HNE in mediating adipocyte differentiation and function in two metabolically distinct obese groups; the insulin sensitive (IS) and the insulin resistant (IR). METHODS: Subcutaneous (SC) adipose tissues were obtained from eighteen clinically well characterized obese premenopausal women undergoing weight reduction surgery. Cellular distribution of 4-HNE in the form of protein adducts was determined by immunohistochemistry in addition to its effect on oxidative stress, cell growth, adipogenic capacity and insulin signaling in preadipocytes derived from the IS and IR participants. RESULTS: 4-HNE was detected in the SC adipose tissue in different cell types with the highest level detected in adipocytes and blood vessels. Short and long-term in vitro treatment of SC preadipocytes with 4-HNE caused inhibition of their growth and increased production of reactive oxygen species (ROS) and antioxidant enzymes. Repeated 4-HNE treatment led to a greater reduction in the adipogenic capacity of preadipocytes from IS subjects compared to IR and caused dephosphorylation of IRS-1 and p70S6K while activating GSK3α/ß and BAD, triggering an IR phenotype. CONCLUSION: These data suggest that 4-HNE-induced oxidative stress plays a role in the regulation of preadipocyte growth, differentiation and insulin signaling and may therefore contribute to adipose tissue metabolic dysfunction associated with insulin resistance.


Asunto(s)
Adipogénesis/efectos de los fármacos , Aldehídos/administración & dosificación , Obesidad/metabolismo , Estrés Oxidativo/efectos de los fármacos , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Adulto , Aldehídos/metabolismo , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Resistencia a la Insulina/genética , Ratones , Obesidad/tratamiento farmacológico , Obesidad/patología , Especies Reactivas de Oxígeno/metabolismo
11.
Front Physiol ; 7: 180, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27252654

RESUMEN

INTRODUCTION: The impact of arthroscopic temperature on joint tissues is poorly understood and it is not known how mesenchymal stem cells (MSCs) respond to the effects of heat generated by the device during the process of arthroscopy assisted experimental cell-based therapy. In the present study, we isolated and phenotypically characterized human bone marrow mesenchymal stem cells (hBMMSCs) from osteoarthritis (OA) patients, and evaluated the effect of arthroscopic heat on cells in suspension and pellet cultures. METHODS: Primary cultures of hBMMSCs were isolated from bone marrow aspirates of OA patients and cultured using DMEM supplemented with 10% FBS and characterized for their stemness. hBMMSCs (1 × 10(6) cells) cultured as single cell suspensions or cell pellets were exposed to an illuminated arthroscope for 10, 20, or 30 min. This was followed by analysis of cellular proliferation and heat shock related gene expression. RESULTS: hBMMSCs were viable and exhibited population doubling, short spindle morphology, MSC related CD surface markers expression and tri-lineage differentiation into adipocytes, chondrocytes and osteoblasts. Chondrogenic and osteogenic differentiation increased collagen production and alkaline phosphatase activity. Exposure of hBMMSCs to an illuminated arthroscope for 10, 20, or 30 min for 72 h decreased metabolic activity of the cells in suspensions (63.27% at 30 min) and increased metabolic activity in cell pellets (62.86% at 10 min and 68.57% at 20 min). hBMMSCs exposed to 37, 45, and 55°C for 120 s demonstrated significant upregulation of BAX, P53, Cyclin A2, Cyclin E1, TNF-α, and HSP70 in cell suspensions compared to cell pellets. CONCLUSIONS: hBMMSC cell pellets are better protected from temperature alterations compared to cell suspensions. Transplantation of hBMMSCs as pellets rather than as cell suspensions to the cartilage defect site would therefore support their viability and may aid enhanced cartilage regeneration.

12.
Diabetologia ; 59(11): 2406-2416, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27342408

RESUMEN

AIMS/HYPOTHESIS: A subset of obese individuals remains insulin sensitive by mechanisms as yet unclear. The hypothesis that maintenance of normal subcutaneous (SC) adipogenesis accounts, at least partially, for this protective phenotype and whether it can be abrogated by chronic exposure to IL-6 was investigated. METHODS: Adipose tissue biopsies were collected from insulin-sensitive (IS) and insulin-resistant (IR) individuals undergoing weight-reduction surgery. Adipocyte size, pre-adipocyte proportion of stromal vascular fraction (SVF)-derived cells, adipogenic capacity and gene expression profiles of isolated pre-adipocytes were determined, along with local in vitro IL-6 secretion. Adipogenic capacity was further assessed in response to exogenous IL-6 application. RESULTS: Despite being equally obese, IR individuals had significantly lower plasma leptin and adiponectin levels and higher IL-6 levels compared with age-matched IS counterparts. Elevated systemic IL-6 in IR individuals was associated with hyperplasia of adipose tissue-derived SVF cells, despite higher frequency of hypertrophied adipocytes. SC pre-adipocytes from these tissues exhibited lower adipogenic capacity accompanied by downregulation of PPARγ (also known as PPARG) and CEBPα (also known as CEBPA) and upregulation of GATA3 expression. Impaired adipogenesis in IR individuals was further associated with increased adipose secretion of IL-6. Treatment of IS-derived SC pre-adipocytes with IL-6 reduced their adipogenic capacity to levels of the IR group. CONCLUSIONS/INTERPRETATION: Obesity-associated insulin resistance is marked by impaired SC adipogenesis, mediated, at least in a subset of individuals, by elevated local levels of IL-6. Understanding the molecular mechanisms underlying reduced adipogenic capacity in IR individuals could help target appropriate therapeutic strategies aimed at those at greatest risk of insulin resistance and type 2 diabetes mellitus.


Asunto(s)
Adipogénesis/fisiología , Resistencia a la Insulina/fisiología , Interleucina-6/metabolismo , Obesidad/metabolismo , Adipocitos/metabolismo , Adipogénesis/genética , Adiponectina/genética , Adiponectina/metabolismo , Adulto , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Humanos , Técnicas In Vitro , Resistencia a la Insulina/genética , Interleucina-6/genética , Masculino , Persona de Mediana Edad , Obesidad/genética , PPAR gamma/genética , PPAR gamma/metabolismo
13.
Stem Cells Dev ; 23(5): 541-54, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24172175

RESUMEN

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


Asunto(s)
Diferenciación Celular/genética , Condrogénesis/genética , Células Madre Mesenquimatosas/citología , Transducción de Señal/genética , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Células de la Médula Ósea/citología , Cartílago/patología , Cartílago/trasplante , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Ingeniería de Tejidos , Factor de Crecimiento Transformador beta3
14.
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
15.
Stem Cell Rev Rep ; 9(3): 350-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21948151

RESUMEN

Transplantation of antigenic-separated stem cells for human cardiovascular diseases such as myocardial infarction needs to be supported by experimental studies that allow refinement of the procedure. In this study we investigated optimising a protocol for the expansion and subsequent differentiation of human umbilical cord blood (HUCB) derived CD133(+) stem cells into a cardiomyocyte-like lineage. CD133(+) cells from HUCB were selected first by immunomagnetic separation and their purity was confirmed by flow cytometry analysis. For expansion and differentiation we developed a novel culture medium recipe that involves sequential signalling factors. Briefly, CD133(+) cells were expanded for 6 days under optimal serum-free conditions in combination with fibronectin and assessed by microscopy and AlamarBlue proliferation assay. Expanded CD133(+) cells were then plated in a cardiac differentiation promoting medium and cultured up to 4 weeks. With this protocol HUCB-CD133(+) cells can be regularly expanded in serum-free medium to obtain recovery and growth in vitro up to 6 folds. The addition of recombinant human thrombopoietin to the remaining factors of the expanding medium was associated with larger cell expansion. Expanded UCB CD133(+) cells showed a cardiomyocyte-like phenotype following differentiation in vitro through expressing intracellular cardiac specific markers including cardiac-specific α-actin, myosin heavy chain and troponin I. This change in phenotype was associated with the expression of cardiac-specific transcription factors Gata-4 and MEF2C. In addition, the change in phenotype was associated with an upregulation of nuclear receptor transcription factors including PPAR α, PPARγ, RXR α and RXRß. We believe our protocol represents a significant advancement and overcome the technical hurdle of deriving cardiomyogenic-like cells from HUCB CD133(+) stem cells. In addition, it has the required attributes of simplicity and consistency. This will permit more robust manipulation of these cells towards better engraftment and repair in patients with myocardial infarction.


Asunto(s)
Antígenos CD/metabolismo , Transdiferenciación Celular , Sangre Fetal/citología , Células Madre Fetales/metabolismo , Glicoproteínas/metabolismo , Miocitos Cardíacos/metabolismo , Péptidos/metabolismo , Antígeno AC133 , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Citometría de Flujo , Humanos , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Receptor alfa X Retinoide/metabolismo , Receptor beta X Retinoide/metabolismo , Trombopoyetina
16.
J Transl Med ; 10: 199, 2012 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-23006493

RESUMEN

BACKGROUND: Glucose-6-phosphate dehydrogenase (G6PD, EC 1.1.1.49) deficiency is caused by one or more mutations in the G6PD gene on chromosome X. An association between enzyme levels and gene haplotypes remains to be established. METHODS: In this study, we determined G6PD enzyme levels and sequenced the coding region, including the intron-exon boundaries, in a group of individuals (163 males and 86 females) who were referred to the clinic with suspected G6PD deficiency. The sequence data were analysed by physical linkage analysis and PHASE haplotype reconstruction. RESULTS: All previously reported G6PD missense changes, including the AURES, MEDITERRANEAN, A-, SIBARI, VIANGCHAN and ANANT, were identified in our cohort. The AURES mutation (p.Ile48Thr) was the most common variant in the cohort (30% in males patients) followed by the Mediterranean variant (p.Ser188Phe) detectable in 17.79% in male patients. Variant forms of the A- mutation (p.Val68Met, p.Asn126Asp or a combination of both) were detectable in 15.33% of the male patients. However, unique to this study, several of such mutations co-existed in the same patient as shown by physical linkage in males or PHASE haplotype reconstruction in females. Based on 6 non-synonymous variants of G6PD, 13 different haplotypes (13 in males, 8 in females) were identified. Five of these were previously unreported (Jeddah A, B, C, D and E) and were defined by previously unreported combinations of extant mutations where patients harbouring these haplotypes exhibited severe G6PD deficiency. CONCLUSIONS: Our findings will help design a focused population screening approach and provide better management for G6PD deficiency patients.


Asunto(s)
Glucosafosfato Deshidrogenasa/genética , Haplotipos , Secuencia de Bases , Estudios de Cohortes , Cartilla de ADN , Femenino , Humanos , Masculino , Mutación , Reacción en Cadena de la Polimerasa , Índice de Severidad de la Enfermedad
17.
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
18.
Biomaterials ; 31(9): 2583-91, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20053438

RESUMEN

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


Asunto(s)
Cartílago/patología , Colágeno/farmacología , Implantes Experimentales , Meniscos Tibiales/patología , Células Madre Mesenquimatosas/citología , Andamios del Tejido/química , Cicatrización de Heridas/efectos de los fármacos , Animales , Cartílago/efectos de los fármacos , Bovinos , Diferenciación Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Humanos , Fenómenos Mecánicos/efectos de los fármacos , Membranas Artificiales , Meniscos Tibiales/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ovinos
19.
Biomaterials ; 30(26): 4277-86, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19539365

RESUMEN

The integration of implanted cartilage is a major challenge for the success of tissue engineering protocols. We hypothesize that in order for effective cartilage integration to take place, matrix-free chondrocytes must be induced to migrate between the two tissue surfaces. A chondrocyte/collagen-scaffold implant system was developed as a method of delivering dividing cells at the interface between two cartilage surfaces. Chondrocytes were isolated from bovine nasal septum and seeded onto both surfaces of a collagen membrane to create the chondrocyte/collagen-scaffold implant. A model of two cartilage discs and the chondrocyte/collagen-scaffold sandwiched in between was used to effect integration in vitro. The resulting tissue was analysed histologically and biomechanically. The cartilage-implant-cartilage sandwich appeared macroscopically as one continuous piece of tissue at the end of 40 day cultures. Histological analysis showed tissue continuum across the cartilage-scaffold interface. The integration was dependent on both cells and scaffold. Fluorescent labeling of implanted chondrocytes demonstrated that these cells invade the surrounding mature tissue and drive a remodelling of the extracellular matrix. Using cell-free scaffolds we also demonstrated that some chondrocytes migrated from the natural cartilage into the collagen scaffold. Quantification of integration levels using a histomorphometric repair index showed that the chondrocyte/collagen-scaffold implant achieved the highest repair index compared to controls, reflected functionally through increased tensile strength. In conclusion, cartilage integration can be achieved using a chondrocyte/collagen-scaffold implant that permits controlled delivery of chondrocytes to both host and graft mature cartilage tissues. This approach has the potential to be used therapeutically for implantation of engineered tissue.


Asunto(s)
Cartílago/metabolismo , Condrocitos/metabolismo , Colágeno/metabolismo , Implantes Experimentales , Oseointegración , Andamios del Tejido , Animales , Cartílago/citología , Bovinos , Movimiento Celular , Condrocitos/citología , Matriz Extracelular/metabolismo , Resistencia a la Tracción
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...