Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Carbohydr Polym ; 340: 122319, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-38858013

RESUMEN

The survival rate of mesenchymal stem cells (MSC), a crucial factor in tissue engineering, is highly dependent on glucose supply. The purpose of this paper is to study the potential of starch foams as glucose suppliers. It is investigated through in vitro hydrolysis by amyloglucosidase in conditions that respect physiological constraints (37 °C and pH 7.4), including a duration of 21 days, and no stirring. Nine extruded starch foams with amylose contents ranging from 0 to 74 %, with various cell wall thicknesses (50 to 300 µm), and different crystallinities (0-30 %) were hydrolysed. These kinetics were fitted by a model which shows that the maximum rate of hydrolysis varies from 7 to 100 %, and which allows the rate of hydrolysis at 21 days to be calculated precisely. The results reveal the major role of amylose in glucose delivery kinetics, and the secondary roles of crystallinity and cell wall thickness of the foams. Additional hydrolysis of starch films revealed that thickness positively influences the amylose chain reorganisation during hydrolysis, which, in slows down and limits glucose delivery. A simple glucose delivery kinetics analysis procedure is proposed to select samples for testing as MSC glucose suppliers.


Asunto(s)
Amilosa , Materiales Biocompatibles , Glucosa , Células Madre Mesenquimatosas , Almidón , Hidrólisis , Glucosa/química , Almidón/química , Materiales Biocompatibles/química , Amilosa/química , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Cinética , Glucano 1,4-alfa-Glucosidasa/metabolismo , Glucano 1,4-alfa-Glucosidasa/química
2.
Bone Joint Res ; 6(4): 208-215, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28408376

RESUMEN

OBJECTIVES: To compare the therapeutic potential of tissue-engineered constructs (TECs) combining mesenchymal stem cells (MSCs) and coral granules from either Acropora or Porites to repair large bone defects. MATERIALS AND METHODS: Bone marrow-derived, autologous MSCs were seeded on Acropora or Porites coral granules in a perfusion bioreactor. Acropora-TECs (n = 7), Porites-TECs (n = 6) and bone autografts (n = 2) were then implanted into 25 mm long metatarsal diaphyseal defects in sheep. Bimonthly radiographic follow-up was completed until killing four months post-operatively. Explants were subsequently processed for microCT and histology to assess bone formation and coral bioresorption. Statistical analyses comprised Mann-Whitney, t-test and Kruskal-Wallis tests. Data were expressed as mean and standard deviation. RESULTS: A two-fold increaseof newly formed bone volume was observed for Acropora-TECs when compared with Porites-TECs (14 sd 1089 mm3versus 782 sd 507 mm3; p = 0.09). Bone union was consistent with autograft (1960 sd 518 mm3). The kinetics of bioresorption and bioresorption rates at four months were different for Acropora-TECs and Porites-TECs (81% sd 5% versus 94% sd 6%; p = 0.04). In comparing the defects that healed with those that did not, we observed that, when major bioresorption of coral at two months occurs and a scaffold material bioresorption rate superior to 90% at four months is achieved, bone nonunion consistently occurred using coral-based TECs. DISCUSSION: Bone regeneration in critical-size defects could be obtained with full bioresorption of the scaffold using coral-based TECs in a large animal model. The superior performance of Acropora-TECs brings us closer to a clinical application, probably because of more suitable bioresorption kinetics. However, nonunion still occurred in nearly half of the bone defects.Cite this article: A. Decambron, M. Manassero, M. Bensidhoum, B. Lecuelle, D. Logeart-Avramoglou, H. Petite, V. Viateau. A comparative study of tissue-engineered constructs from Acropora and Porites coral in a large animal bone defect model. Bone Joint Res 2017;6:208-215. DOI: 10.1302/2046-3758.64.BJR-2016-0236.R1.

3.
Eur Cell Mater ; 31: 160-73, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26895242

RESUMEN

The effects of mechanical stimuli to which cells are exposed in vivo are, at best, incompletely understood; in this respect, gene-level information regarding cell functions which are pertinent to new tissue formation is of special interest and importance in applications such as tissue engineering and tissue regeneration. Motivated by this need, the present study investigated the early responses of human mesenchymal stem cells (hMSCs) to intermittent shear stress (ISS) and to cyclic hydrostatic pressure (CHP) simulating some aspects of the biological milieu in which these cells exist in vivo. Production of nitric oxide (NO) and mRNA expression of several known mechanosensitive genes as well as ERK1/2 activation in the hMSC response to the two mechanical stimuli tested were monitored and compared. NO production depended on the type of the mechanical stimulus to which the hMSCs were exposed and was significantly higher after exposure to ISS than to CHP. At the conditions of NO peak release (i.e., at 0.7 Pa for ISS and 50,000 Pa for CHP), ISS was more effective than CHP in up-regulating mechanosensitive genes. ERK1/2 was activated by ISS but not by CHP. The present study is the first to report that PGTS2, IER3, EGR1, IGF1, IGFBP1, ITGB1, VEGFA and FGF2 are involved in the response of hMSCs to ISS. These findings establish that, of the two mechanical stimuli tested, ISS is more effective than CHP in triggering expression of genes from hMSCs which are bioactive and pertinent to several cell functions (such as cell differentiation and release of specific growth factors and cytokines) and also to tissue-related processes such as wound healing.


Asunto(s)
Presión Hidrostática/efectos adversos , Mecanotransducción Celular/fisiología , Células Madre Mesenquimatosas/fisiología , Estrés Fisiológico/fisiología , Células de la Médula Ósea/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Óxido Nítrico/biosíntesis , ARN Mensajero/biosíntesis , Estrés Mecánico , Ingeniería de Tejidos/métodos
4.
Orthop Traumatol Surg Res ; 102(2): 189-95, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26775085

RESUMEN

BACKGROUND: Tissue engineering strategies include both cell-based and cell homing therapies. Ligamentous tissues are highly specialized and constitute vital components of the musculoskeletal system. Their damage causes significant morbidity and loss in function. HYPOTHESIS: The aim of this study is to analyze tendinous graft integration, cell repopulation and ligamentization by using GFP+/- allografts in GFP+/- transgenic New Zealand white (NZW) rabbits. MATERIAL AND METHODS: Graft implantation was designed to closely mimic anterior cruciate ligament (ACL) repair surgery. Allografts were implanted in 8 NZW rabbits and assessed at 5 days, 3 weeks and 6 weeks through: (1) arthroCT imaging, (2) morphological analysis of the transplanted allograft, (3) histological analysis, (4) collagen type I immunochemistry, and (5) GFP cell tracking. Collagen remodeling was appreciated at 3 and 6 weeks. RESULTS: Graft repopulation with host cells, chondrocyte-like cells at the tendon-bone interface and graft corticalization in the bone tunnels were noticed at 3 weeks. By contrast we noticed a central necrosis aspect in the allografts intra-articularly at 6 weeks with a cell migration towards the graft edge near the synovium. DISCUSSION: Our study has served to gain a better understanding of tendinous allograft bone integration, ligamentization and allograft repopulation. We believe that both cell-based therapies and cell homing therapies are beneficial in ligament tissue engineering. Future studies may elucidate whether cell repopulation occurs with pre-differentiated or progenitor cells. We believe that both cell-based therapies and cell homing therapies are beneficial in ligament tissue engineering. LEVEL OF EVIDENCE: Level V (animal study).


Asunto(s)
Aloinjertos/patología , Aloinjertos/fisiología , Reconstrucción del Ligamento Cruzado Anterior , Tendones/patología , Tendones/trasplante , Aloinjertos/citología , Animales , Huesos/fisiología , Movimiento Celular , Rastreo Celular , Colágeno Tipo I/análisis , Colágeno Tipo I/ultraestructura , Necrosis , Conejos , Tendones/citología , Tendones/fisiología , Factores de Tiempo , Ingeniería de Tejidos
5.
J Mater Sci Mater Med ; 26(1): 5339, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25578693

RESUMEN

Although bone substitutes associated with platelet concentrates are widely used to vertically reconstruct alveolar ridges, their respective and specific contribution remain controversial. The aim of this study was to evaluate the benefit of using either biphasic calcium phosphate (BCP) or demineralised bovine bone mineral (DBBM) alone or with autologous platelet lysate (APL) in vertical bone regeneration. The study involved fourteen New Zealand rabbits. Autologous APL was prepared by freeze-thawing from a platelet suspension (10(9) platelets/ml). Four CP titanium (cpTi) cylinders were fixed to each calvarium; one cylinder was empty, one was filled with APL alone and the others were filled either with BCP or BCP + APL or DBBM or DBBM + APL. New bone formation and biomaterial resorption were evaluated using non-demineralised histology and histomorphometry. After 6 weeks, new bone formation was observed in all cylinders. The newly formed bone in the cylinders filled with APL alone, DBBM and BCP was significantly increased by (0.6-, 2.5- and 3.3-fold, respectively) (P < 0.0001) compared to results obtained with the empty cylinders. Vertical bone height in the cylinders filled with BCP was greater to that observed with DBBM. The residual material in the cylinders filled with BCP was significantly (P < 0.0001) lower (0.35-fold) than that with DBBM. Both newly formed bone and residual material in the cylinders filled with BCP + APL or DBBM + APL were similar to those filled with either BCP or DBBM, respectively. This study provided evidence that APL alone, as well as DBBM and BCP, have a beneficial effect on vertical bone formation and remodelling. APL associated with either DBBM or BCP did not provide additional benefits.


Asunto(s)
Plaquetas , Regeneración Ósea , Fosfatos de Calcio/metabolismo , Cráneo/patología , Animales , Calcificación Fisiológica , Bovinos , Conejos
6.
J Mater Sci Mater Med ; 24(7): 1745-54, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23625318

RESUMEN

Titanium surface modifications to simultaneously prevent bacterial adhesion but promote bone-cell functions could be highly beneficial for improving implant osseointegration. In the present in vitro study, the effect of sulfonate groups on titanium surfaces was investigated with respect to both S. aureus adhesion and osteoblast functions pertinent to new bone formation. Commercial pure titanium (cpTi) squares were oxydized (Tiox), grafted with poly(sodium styrene sulfonate) groups (Tigraft) by covalent bonding using radical polymerization, and were characterized by infrared spectroscopy (HATR-FTIR) and colorimetry. Bacterial adhesion study showed that Tigraft exhibited high inhibition of S. aureus adhesion S at levels >90 %, when compared to cpTi (P < 0.05). In contrast osteoblasts adhesion was similar on all three titanium surfaces. While the kinetics of cell proliferation were similar on the three titanium surfaces, Alkaline phosphatase-specific activity of osteoblasts cultured on Tigraft surfaces was twofold higher than that observed on either on Tiox or cpTi surfaces (P < 0.01). More importantly, the amount and the distribution of calcium-containing nodules was different. The total area covered by calcium-containing nodules was 2.2-fold higher on the Tigraft as compared to either Tiox or cpTi surfaces (P < 0.01). These results provide evidence that poly(sodium styrene sulfonate) groups grafting on cpTi simultaneously inhibits bacteria adhesion but promote osteoblast function pertinent to new bone formation. Such modified titanium surfaces offer a promising strategy for preventing biofilm-related infections and enhancing osteointegration of implants in orthopaedic and dental applications.


Asunto(s)
Adhesión Bacteriana/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Materiales Biocompatibles Revestidos/farmacología , Osteoblastos/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Animales , Células Cultivadas , Materiales Biocompatibles Revestidos/síntesis química , Materiales Biocompatibles Revestidos/química , Embrión de Mamíferos , Ensayo de Materiales , Pruebas de Sensibilidad Microbiana , Oseointegración/efectos de los fármacos , Osteoblastos/fisiología , Osteogénesis/efectos de los fármacos , Polímeros/síntesis química , Polímeros/química , Polímeros/farmacología , Poliestirenos/síntesis química , Poliestirenos/química , Poliestirenos/farmacología , Ratas , Ratas Wistar , Sodio/química , Sodio/farmacología , Staphylococcus aureus/fisiología , Compuestos de Sulfonio/síntesis química , Compuestos de Sulfonio/química , Compuestos de Sulfonio/farmacología , Titanio/química , Titanio/farmacología
7.
Eur Cell Mater ; 24: 18-28, 2012 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-22777950

RESUMEN

Transplantation of mesenchymal stem cells (MSCs) with electrotransferred bone morphogenetic protein-2 (BMP-2) transgene is an attractive therapeutic modality for the treatment of large bone defects: it provides both stem cells with the ability to form bone and an effective bone inducer while avoiding viral gene transfer. The objective of the present study was to determine the influence of the promoter driving the human BMP-2 gene on the level and duration of BMP-2 expression after transgene electrotransfer into rat MSCs. Cytomegalovirus, elongation factor-1α, glyceraldehyde 3-phosphate dehydrogenase, and beta-actin promoters resulted in a BMP-2 secretion rate increase of 11-, 78-, 66- and 36-fold over respective controls, respectively. In contrast, the osteocalcin promoter had predictable weak activity in undifferentiated MSCs but induced the strongest BMP-2 secretion rates in osteoblastically-differentiated MSCs. Regardless of the promoter driving the transgene, a plateau of maximal BMP-2 secretion persisted for at least 21 d after the hBMP-2 gene electrotransfer. The present study demonstrates the feasibility of gene electrotransfer for efficient BMP-2 transgene delivery into MSCs and for a three-week sustained BMP-2 expression. It also provides the first in vitro evidence for a safe alternative to viral methods that permit efficient BMP-2 gene delivery and expression in MSCs but raise safety concerns that are critical when considering clinical applications.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Expresión Génica , Operón Lac , Osteocalcina/genética , Animales , Proteína Morfogenética Ósea 2/metabolismo , Huesos/citología , Huesos/fisiología , Diferenciación Celular , Células Cultivadas , Electroporación , Técnicas de Transferencia de Gen , Humanos , Masculino , Células Madre Mesenquimatosas , Ratones , Osteocalcina/metabolismo , Plásmidos , Regiones Promotoras Genéticas , Ratas , Ratas Endogámicas Lew , Factores de Tiempo , Transgenes
8.
Osteoporos Int ; 22(6): 2017-21, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21523397

RESUMEN

Bioengineered bone scaffolds are intended for use in large bone defects. Successful bone constructs should stimulate and support both the onset and the continuance of bone ingrowth. In an attempt to improve their performance and to compete with the one of autologous bone grafts, a growing symbiosis at the biological and material level is required. Recent advances have been made to further exploit the osteogenic potential of MSCs in scaffold development. Current research encompasses new strategies for reducing cell death after implantation and the manufacturing of tailored, instructive scaffolds.


Asunto(s)
Sustitutos de Huesos/uso terapéutico , Ingeniería de Tejidos/métodos , Apoptosis , Trasplante Óseo/métodos , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/patología , Osteogénesis/fisiología , Andamios del Tejido
9.
J Cell Mol Med ; 15(7): 1505-14, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20716129

RESUMEN

Use of mesenchymal stem cells (MSCs) has emerged as a potential new treatment for various diseases but has generated marginally successful results. A consistent finding of most studies is massive death of transplanted cells. The present study examined the respective roles of glucose and continuous severe hypoxia on MSC viability and function with respect to bone tissue engineering. We hereby demonstrate for the first time that MSCs survive exposure to long-term (12 days), severe (pO(2) < 1.5 mmHg) hypoxia, provided glucose is available. To this end, an in vitro model that mimics the hypoxic environment and cell-driven metabolic changes encountered by grafted sheep cells was established. In this model, the hallmarks of hypoxia (low pO(2) , hypoxia inducible factor-1α expression and anaerobic metabolism) were present. When conditions switched from hypoxic (low pO(2) ) to ischemic (low pO(2) and glucose depletion), MSCs exhibited shrinking, decreased cell viability and ATP content due to complete exhaustion of glucose at day 6; these results provided evidence that ischemia led to the observed massive cell death. Moreover, MSCs exposed to severe, continuous hypoxia, but without any glucose shortage, remained viable and maintained both their in vitro proliferative ability after simulation with blood reperfusion at day 12 and their in vivo osteogenic ability. These findings challenge the traditional view according to which severe hypoxia per se is responsible for the massive MSC death observed upon transplantation of these cells and provide evidence that MSCs are able to withstand exposure to severe, continuous hypoxia provided that a glucose supply is available.


Asunto(s)
Hipoxia de la Célula/fisiología , Supervivencia Celular/fisiología , Glucosa/metabolismo , Células Madre Mesenquimatosas/fisiología , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isquemia/metabolismo , Ácido Láctico/metabolismo , Células Madre Mesenquimatosas/citología , Oxígeno/metabolismo , Ovinos , Ingeniería de Tejidos , Andamios del Tejido
10.
Chir Main ; 29(5): 307-14, 2010 Oct.
Artículo en Francés | MEDLINE | ID: mdl-20728395

RESUMEN

A prospective study is reported concerning 11 cases of bone defect of the hand and wrist treated by the induced membrane technique. Ten men and one woman with an average age of 49 yrs (17-72) sustained a high-energy trauma with severe mutilation of digit and hand but with intact pulp. Eight cases of open finger fractures with composite loss of substance and three cases of bone and joint infection (thumb, wrist, fifth finger) were included. All cases were treated by the induced membrane technique which consists in stable fixation, flap if necessary, and in filling the bone defect by a cement methyl methacrylate polymere (PMMA) spacer. A secondary procedure at two months is needed where the cement is removed and the void is filled by cancellous bone. The key point of this induced membrane technique is to respect the foreign body membrane which formed around the cement spacer creating a biologic chamber. Bone union was evaluated prospectively by X-ray and CT scan by a surgeon not involved in the treatment. Failure was defined as non-union at one year, or uncontrolled sepsis at one month. Two cases failed to achieve bone union. No septic complications occurred and all septic cases were controlled. In nine cases, bone union was achieved within four months (three to 12). Evidence of osteoid formation was determined by a bone biopsy in one case. Masquelet first reported 35 cases of large tibial non-union defects treated by the induced membrane technique. The cement spacer promotes foreign body membrane induction constituting a biological chamber. Works on animal models reported by Pellissier and Viateau demonstrated membrane properties: secretion of growths factors (VEGF, TGF beta1, BMP2) and osteoinductive cellular activity. The induced membrane seems to mimic a neoperiosteum. This technique is useful in emergency or septic conditions where bone defects cannot be treated by shortening. It avoids microsurgery and is limited by availability of cancellous bone.


Asunto(s)
Cementos para Huesos/uso terapéutico , Fracturas Abiertas/cirugía , Traumatismos de la Mano/cirugía , Membranas Artificiales , Polimetil Metacrilato/uso terapéutico , Traumatismos de la Muñeca/cirugía , Adolescente , Adulto , Anciano , Urgencias Médicas , Femenino , Fracturas Abiertas/diagnóstico por imagen , Traumatismos de la Mano/diagnóstico por imagen , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Radiografía , Colgajos Quirúrgicos , Resultado del Tratamiento , Traumatismos de la Muñeca/diagnóstico por imagen
11.
Gene Ther ; 15(7): 537-44, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18256695

RESUMEN

Gene electrotransfer is an efficient and reproducible nonviral gene transfer technique useful for the nonpermanent expression of therapeutic transgenes. The present study established optimal conditions for the electrotransfer of reporter genes into mesenchymal stem cells (MSCs) isolated from rat bone marrow by their selective adherence to tissue-culture plasticware. The electrotransfer of the lacZ reporter gene was optimized by adjusting the pulse electric field intensity, electric pulse type, electropulsation buffer conductivity and electroporation temperature. LacZ electrotransfection into MSCs was optimal at 1500 V cm(-1) with pre-incubation in Spinner's minimum essential medium buffer at 22 degrees C. Under these conditions beta-galactosidase expression was achieved in 29+/-3% of adherent cells 48 h post transfection. The kinetics of beta-galactosidase activity revealed maintenance of beta-galactosidase production for at least 10 days. Moreover, electroporation did not affect the MSC potential for multidifferentiation; electroporated MSCs differentiated into osteoblastic, adipogenic and chondrogenic lineages to the same extent as cells that were not exposed to electric pulses. Thus, this study demonstrates the feasibility of efficient transgene electrotransfer into MSCs while preserving cell viability and multipotency.


Asunto(s)
Electroporación/métodos , Terapia Genética/métodos , Células Madre Mesenquimatosas/metabolismo , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Expresión Génica , Operón Lac , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratas , Ratas Endogámicas Lew , Temperatura , Factores de Tiempo , Transfección/métodos , Transgenes , beta-Galactosidasa/genética
12.
Tissue Eng ; 13(1): 87-99, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17518583

RESUMEN

Fabrication of implantable cartilaginous structures that could be secured in the joint defect could provide an alternative therapeutic approach to prosthetic joint replacement. Herein we explored the possibility of using biodegradable hydrogels in combination with a polyglycolic acid (PGA) scaffold to provide an environment propitious to mesenchymal stem cells (MSCs) chondrogenic differentiation. We examined the influence of type I collagen gel and alginate combined with PGA meshes on the extracellular matrix composition of tissue-engineered transplants. MSCs were isolated from young rabbits, expanded in monolayers, suspended in each hydrogel, and loaded on PGA scaffolds. All constructs (n=48) were cultured in serum-free medium containing transforming growth factor beta-1, under dynamic conditions in specially designed bioreactors for 3-6 weeks. All cell-polymer constructs had a white, shiny aspect, and retained their initial size and shape over the culture period. Their thickness increased substantially over time, and no shrinkage was observed. All specimens developed a hyalin-like extracellular matrix containing glycosaminoglycans (GAGs) and type II collagen, but significant differences were observed among the three different groups. In PGA/MSCs and collagen-PGA/MSCs constructs, the cell growth phase and the chondrogenic differentiation phase of MSCs occurred during the first 3 weeks. In alginate-PGA/MSCs constructs, cells remained round in the hydrogel and cartilage extracellular matrix deposition was delayed. However, at 6 weeks, alginate-PGA/MSCs constructs exhibited higher contents of GAGs and lower contents of type I collagen. These results suggest that the implied time for the transplantation of in vitro engineered constructs depends, among other factors, on the nature of the scaffold envisioned. In this study, we demonstrated that the use of a composite hydrogel-PGA scaffold supported the in vitro growth of implantable cartilaginous structures cultured in a bioreactor system.


Asunto(s)
Materiales Biocompatibles , Cartílago Hialino/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Ingeniería de Tejidos/métodos , Alginatos/ultraestructura , Animales , Materiales Biocompatibles/síntesis química , Reactores Biológicos , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Células de la Médula Ósea/ultraestructura , Adhesión Celular/fisiología , Técnicas de Cultivo de Célula , Colágeno Tipo I/síntesis química , Colágeno Tipo I/ultraestructura , Colágeno Tipo II/síntesis química , Colágeno Tipo II/ultraestructura , Ácido Glucurónico/fisiología , Ácidos Hexurónicos , Cartílago Hialino/fisiología , Cartílago Hialino/ultraestructura , Hidrogeles , Masculino , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/ultraestructura , Microscopía Fluorescente , Ácido Poliglicólico , Conejos
13.
J Histochem Cytochem ; 55(3): 255-62, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17101724

RESUMEN

Considerable research has been focused on the use of bone marrow-derived mesenchymal stem cells (MSCs) for the repair of non-unions and bone defects. To date, the question of whether transplanted MSCs survive and engraft within newly formed tissue remains unresolved. The development of an easy and reliable method that would allow cell fate monitoring in transplant recipients is a pressing concern for the field of tissue engineering. To demonstrate the presence of transplanted cells in newly formed bone, we established a xenograft nude rat model allowing the detection of murine LacZ MSCs in vivo. MSCs were isolated from transgenic lacZ mice, seeded onto bioabsorbable collagen sponges, and transplanted to repair a calvarial defect in nude rats. As a preliminary step, the histological procedure was adapted to optimize the detection of LacZ cells in bone tissue embedded in methylmethacrylate (MMA). Four fixatives and four fixation times were evaluated. Among all the fixatives tested, 2% formaldehyde/0.2% glutaraldehyde at 4C for 4 days gave the best results for X-gal staining at pH 7.4 on both cell cultures and bone explants. All fixatives were effective for immunodetection of beta-gal. In the chimeric LacZ/nude rat animal model, MSCs were detected in vivo for up to 4 weeks after implantation and contributed to the repair and the neovascularization of the bone defect. LacZ is a suitable phenotypic marker to track MSCs in skeletal tissues embedded in MMA.


Asunto(s)
Huesos/citología , Operón Lac , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Metilmetacrilato , Adhesión del Tejido , Animales , Colágeno , Fijadores , Genes Reporteros , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Transgénicos , Ratas , Ratas Desnudas , Factores de Tiempo , Fijación del Tejido , Trasplante Heterólogo , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
14.
Anal Biochem ; 349(1): 78-86, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16307714

RESUMEN

Bone morphogenetic proteins (BMPs) control cell fate by regulating gene expression, especially inhibitor of differentiation (Id) genes. This property has been exploited to create a highly sensitive assay for quantification of active BMP. Embryonic mouse cells (C3H10T1/2) were stably transfected with an expression construct (BRE-Luc) containing a BMP-responsive element fused to the firefly luciferase reporter gene. BRE results from a multimerization of distinct sequences elements from a mouse Id1 promoter [15]. The addition of BMP-2 (0.5-100ng/ml) to the transfectants resulted in a dose-dependent increase in luciferase activity in the cell lysates. This new assay was 100-fold more sensitive than the classical alkaline phosphatase (ALP) activity assay (0.5-1 vs. 50-100ng/ml, respectively) as well as much more rapid (24h vs. 3-6 days, respectively, of BMP treatment). This new assay is specific to BMPs (BMP-2, BMP-4, and BMP7) as evidenced by its relative insensitivity to TGFbeta1, bFGF, and VEGF. Because of its BMP specificity, this rapid, sensitive, nonradioactive, and easily performed assay could be used in monitoring the biological activity of BMP and, eventually, as a cell-based screening assay to identify and evaluate molecules that modulate BMP signaling in cells.


Asunto(s)
Proteínas Morfogenéticas Óseas/análisis , Luciferasas/genética , Regiones Promotoras Genéticas , Transfección , Animales , Proteínas Morfogenéticas Óseas/genética , Línea Celular , Células Clonales , Humanos , Ratones , Ratones Endogámicos C3H
15.
Tissue Eng ; 11(5-6): 814-24, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15998221

RESUMEN

Large bone defects are still a challenge to orthopedic surgeons. In this study, a massive bone defect with a clinically relevant volume was efficiently reconstructed by transplanting an engineered bone in which mesenchymal stem cells (MSCs) expanded in autologous serum (AS) were combined with a porous scaffold. In the first step, we established that the way in which the MSCs are distributed over the scaffold affects the ultimate bone-forming ability of the transplant: constructs consisting of a natural coral scaffold and a pseudo-periosteal layer of MSCs surrounding the implant (coral-MSC3D) formed significantly more bone than constructs in which the MSCs were distributed throughout the implant (p = 0.01). However, bone healing occurred in only one sheep, owing to the high resorption rate of natural coral scaffold. To overcome this problem, constructs in which MSCs were combined with a porous coralline-based hydroxyapatite (CHA) scaffold having the same architecture as natural coral but a lower resorption rate were prepared. After their implantation, these constructs were found to have the same osteogenic potential as autologous bone grafts in terms of the amount of newly formed bone present at 4 months (p = 0.89) and to have been completely replaced by newly formed, structurally competent bone within 14 months. Nevertheless, although the rate of bone healing was strikingly improved when CHA-MSC3D constructs were used (five of seven animals healed) as compared with the coral-MSC3D construct (one of seven healed), it was still less satisfactory than that obtained with autografts (five of five healed).


Asunto(s)
Sustitutos de Huesos , Huesos Metatarsianos , Ingeniería de Tejidos , Animales , Antozoos , Regeneración Ósea/fisiología , Durapatita , Células Madre Mesenquimatosas , Huesos Metatarsianos/cirugía , Prótesis e Implantes , Ovinos
16.
J Cell Mol Med ; 9(1): 72-84, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15784166

RESUMEN

Repair of large bone defects is still a challenge for the orthopaedic, reconstructive and maxillo-facial surgeon. Availability of pluripotent stem cells from either autologous or allogenic sources and the potential of inducing the osteogenic phenotype is motivating exploration and development of custom-tailored materials known as "bioengineered bone constructs". In such cases, the clinical scenario involves either expansion of stem cells in monolayer and loading them into a porous scaffold prior to surgery or direct cell expansion within the scaffold, and implanting this novel construct back into the donor patient. In this review, we delineate, from an engineering perspective, the progress that has been made to date and the challenges remaining in successfully translating this promising (but not yet definitively established) approach from bench to the bed site.


Asunto(s)
Huesos/citología , Ingeniería de Tejidos/métodos , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/uso terapéutico , Trasplante de Médula Ósea , Sustitutos de Huesos/química , Sustitutos de Huesos/uso terapéutico , Huesos/cirugía , Humanos , Osteogénesis , Células Madre
18.
Pathol Biol (Paris) ; 53(3): 142-8, 2005 Apr.
Artículo en Francés | MEDLINE | ID: mdl-15781372

RESUMEN

Stem cell therapy of skeletal tissues involves the transplantation of stem cells to the tissues that have been damaged by injury or disease. Although these cells can be derived from embryos, the preferred source of skeletal stem cells is the bone marrow as it contains adult stem cells that can be easily driven towards a bone phenotype. More recently, cells with similar potentialities have also been derived from adipose tissue, muscle, or blood. A biomaterial (ceramics or polymers) is often required as a scaffold to promote cell adhesion, proliferation and differentiation as well as encourage vascular invasion and ultimately new bone formation. The first clinical studies are encouraging and suggests that stem cell therapy could be a prime method for bone reconstruction.


Asunto(s)
Mesodermo/citología , Ortopedia/tendencias , Trasplante de Células Madre/métodos , Adulto , División Celular , Humanos , Células Madre/citología
19.
Biomaterials ; 24(14): 2497-502, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12695076

RESUMEN

A potential therapy to enhance healing of bone tissue is to deliver isolated mesenchymal stem cells (MSCs) to the site of a lesion to promote bone formation. A key issue within this technology is the development of an injectable system for the delivery of MSCs. Fibrin gel exploits the final stage of the coagulation cascade in which fibrinogen molecules are cleaved by thrombin, convert into fibrin monomers and assembled into fibrils, eventually forming fibers in a three-dimensional network. This gel could have many advantages as a cell delivery vehicle in terms of biocompatibility, biodegradation and hemostasis. The objective of this study was to explore the possibility of using fibrin gel as a delivery system for human MSCs (HMSCs). To this end we have determined the optimal fibrinogen concentrations and thrombin activity for loading HMSCs in vitro into the resultant fibrin gels to obtain cell proliferation. We found that a concentration of 18 mg/ml of fibrinogen and a thrombin activity of 100 IU/ml was optimal for producing fibrin scaffolds that would allow good HMSCs spreading and proliferation. In these conditions, cells were able to proliferate and expressed alkaline phosphatase, a bone marker, in vitro. When implanted in vivo, HMSCs were able to migrate out of the fibrin gel and invade a calcium carbonate based ceramic scaffold suggesting that fibrin gel could serve as a delivery system for HMSCs.


Asunto(s)
Técnicas de Cultivo/métodos , Matriz Extracelular/metabolismo , Fibrina/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cultivo/instrumentación , Matriz Extracelular/química , Humanos , Trasplante de Células Madre Mesenquimatosas/instrumentación , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Persona de Mediana Edad , Trombina/farmacología
20.
J Biomed Mater Res ; 56(3): 361-7, 2001 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-11372053

RESUMEN

Recent advances in the field of tissue engineering have culminated in new tissue substitutes that combine a biomaterial and precursor cells. The effectiveness of these materials is generally assessed in animals, but few studies explore the fate of the transplanted cells in vivo, despite its paramount importance for understanding the function of the engineered tissues. Current methods that use reporter genes or chimeric animals are not always well suited to solving tissue-engineering problems. We therefore developed a new method for irreversible labeling of cells to track their fate in vivo. We used a fluorescent lipophilic probe, CM-Dil, that avidly binds to the cell membrane. Human bone marrow stromal fibroblasts could be labeled with 20 microM CM-Dil in 30 min. The CM-Dil was not cytotoxic and did not affect cell proliferation in vitro. Cells could be monitored for up to 30 days when placed in a coral scaffold and implanted intramuscularly or in a bony site. However, the fluorescence intensity decreased roughly in parallel with the number of cell divisions. This fact needs to be taken into account during the design and interpretation of experiments. We believe that this technique is also of interest for other cell types.


Asunto(s)
Células de la Médula Ósea/citología , Carbocianinas , Fibroblastos/citología , Colorantes Fluorescentes , Movimiento Celular , Trasplante de Células , Células Cultivadas , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...