Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am J Sports Med ; 52(6): 1596-1607, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38581200

RESUMEN

BACKGROUND: Physiological 0.9% saline is commonly used as an irrigation fluid in modern arthroscopy. There is a growing body of evidence that a hyperosmolar saline solution has chondroprotective effects, especially if iatrogenic injury occurs. PURPOSE: To (1) corroborate the superiority of a hyperosmolar saline solution regarding chondrocyte survival after mechanical injury and (2) observe the modulatory response of articular cartilage to osmotic stress and injury. STUDY DESIGN: Controlled laboratory study. METHODS: Osteochondral explants were isolated from bovine stifle joints and exposed to either 0.9% saline (308 mOsm) or hyperosmolar saline (600 mOsm) and then damaged with a sharp dermatome blade to attain a confined full-thickness cartilage injury site, incubated in the same fluids for another 3 hours, and transferred to chondropermissive medium for further culture for 1 week. Chondrocyte survival was assessed by confocal imaging, while the cellular response was evaluated over 1 week by relative gene expression for apoptotic and inflammatory markers and mediator release into the medium. RESULTS: The full-thickness cartilage cut resulted in a confined zone of cell death that mainly affected superficial zone chondrocytes. Injured samples that were exposed to hyperosmolar saline showed less expansion of cell death in both the axial (P < .007) and the coronal (P < .004) plane. There was no progression of cell death during the following week of culture. Histological assessment revealed an intact cartilage matrix and normal chondrocyte morphology. Inflammatory and proapoptotic genes were upregulated on the first days postexposure with a notable downregulation toward day 7. Mediator release into the medium was concentrated on day 3. CONCLUSION: This in vitro cartilage injury model provides further evidence for the chondroprotective effect of a hyperosmolar saline irrigation fluid, as well as novel data on the capability of articular cartilage to quickly regain joint homeostasis after osmotic stress and injury. CLINICAL RELEVANCE: Raising the osmolarity of an irrigating solution may be a simple and safe strategy to protect articular cartilage during arthroscopic surgery.


Asunto(s)
Cartílago Articular , Condrocitos , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/lesiones , Bovinos , Condrocitos/efectos de los fármacos , Presión Osmótica , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Irrigación Terapéutica , Solución Salina
2.
Materials (Basel) ; 16(22)2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-38005143

RESUMEN

Osteochondral (OC) disorders such as osteoarthritis (OA) damage joint cartilage and subchondral bone tissue. To understand the disease, facilitate drug screening, and advance therapeutic development, in vitro models of OC tissue are essential. This study aims to create a bioprinted OC miniature construct that replicates the cartilage and bone compartments. For this purpose, two hydrogels were selected: one composed of gelatin methacrylate (GelMA) blended with nanosized hydroxyapatite (nHAp) and the other consisting of tyramine-modified hyaluronic acid (THA) to mimic bone and cartilage tissue, respectively. We characterized these hydrogels using rheological testing and assessed their cytotoxicity with live-dead assays. Subsequently, human osteoblasts (hOBs) were encapsulated in GelMA-nHAp, while micropellet chondrocytes were incorporated into THA hydrogels for bioprinting the osteochondral construct. After one week of culture, successful OC tissue generation was confirmed through RT-PCR and histology. Notably, GelMA/nHAp hydrogels exhibited a significantly higher storage modulus (G') compared to GelMA alone. Rheological temperature sweeps and printing tests determined an optimal printing temperature of 20 °C, which remained unaffected by the addition of nHAp. Cell encapsulation did not alter the storage modulus, as demonstrated by amplitude sweep tests, in either GelMA/nHAp or THA hydrogels. Cell viability assays using Ca-AM and EthD-1 staining revealed high cell viability in both GelMA/nHAp and THA hydrogels. Furthermore, RT-PCR and histological analysis confirmed the maintenance of osteogenic and chondrogenic properties in GelMA/nHAp and THA hydrogels, respectively. In conclusion, we have developed GelMA-nHAp and THA hydrogels to simulate bone and cartilage components, optimized 3D printing parameters, and ensured cell viability for bioprinting OC constructs.

3.
Mater Today Bio ; 22: 100775, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37674778

RESUMEN

Herein we show an accessible technique based on Faraday waves that assist the rapid assembly of osteoinductive ß-Tricalcium phosphate (ß-TCP) particles as well as human osteoblast pre-assembled in spheroids. The hydrodynamic forces originating at 'seabed' of the assembly chamber can be used to tightly aggregate inorganic and biological entities at packing densities that resemble those of native tissues. Additionally, following a layer-by-layer assembly procedure, centimeter scaled osteoinductive three-dimensional and cellularized constructs have been fabricated. We showed that the intimate connection between biological building blocks is essential in engineering living system able of localized mineral deposition. Our results demonstrate, for the first time, the possibility to obtain three-dimensional cellularized and acellularized anisotropic constructs using Faraday waves.

4.
Biomed Res Int ; 2022: 7638245, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35118158

RESUMEN

Distinctive characteristics of articular cartilage such as avascularity and low chondrocyte conversion rate present numerous challenges for orthopedists. Tissue engineering is a novel approach that ameliorates the regeneration process by exploiting the potential of cells, biodegradable materials, and growth factors. However, problems exist with the use of tissue-engineered construct, the most important of which is scaffold-cartilage integration. Recently, many attempts have been made to address this challenge via manipulation of cellular, material, and biomolecular composition of engineered tissue. Hence, in this review, we highlight strategies that facilitate cartilage-scaffold integration. Recent advances in where efficient integration between a scaffold and native cartilage could be achieved are emphasized, in addition to the positive aspects and remaining problems that will drive future research.


Asunto(s)
Cartílago Articular , Ingeniería de Tejidos , Condrocitos , Regeneración , Andamios del Tejido
5.
Stem Cell Res Ther ; 11(1): 436, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-33036643

RESUMEN

BACKGROUND: Mesenchymal stem cells are a promising cell source for chondrogenic differentiation and have been widely used in several preclinical and clinical studies. However, they are prone to an unwanted differentiation process towards hypertrophy that limits their therapeutic efficacy. Matrix metallopeptidase 13 (MMP-13) is a well-known factor regulated during this undesirable event. MMP-13 is a collagen degrading enzyme, which is also highly expressed in the hypertrophic zone of the growth plate and in OA cartilage. Accordingly, we investigated the effect of MMP-13 inhibition on MSC hypertrophy. METHODS: In this study, 5-bromoindole-2-carboxylic acid (BICA) was used as an inhibitory agent for MMP-13 expression. After identifying its optimal concentration, BICA was mixed into a hydrogel and the release rate was studied. To prepare the ideal hydrogel, chondroitin sulfate (CS) and platelet lysate (PL) were mixed with sodium alginate (Alg) at concentrations selected based on synergistic mechanical and rheometric properties. Then, four hydrogels were prepared by combining alginate (1.5%w/v) and/or CS (1%w/v) and/or PL (20%v/v). The chondrogenic potential and progression to hypertrophy of human bone marrow-derived mesenchymal stem cell (hBM-MSC)-loaded hydrogels were investigated under free swelling and mechanical loading conditions, in the presence and absence of BICA. RESULTS: Viability of hBM-MSCs seeded in the four hydrogels was similar. qRT-PCR revealed that BICA could successfully inhibit MMP-13 expression, which led to an inhibition of Coll X and induction of Coll-II, in both free swelling and loading conditions. The GAG deposition was higher in the group combining BICA and mechanical stimulation. CONCLUSIONS: It is concluded that BICA inhibition of MMP-13 reduces MSC hypertrophy during chondrogenesis.


Asunto(s)
Diferenciación Celular , Condrogénesis , Hidrogeles , Inhibidores de la Metaloproteinasa de la Matriz , Alginatos , Células Cultivadas , Condrocitos , Sulfatos de Condroitina/farmacología , Humanos , Hipertrofia , Metaloproteinasa 13 de la Matriz/genética , Células Madre Mesenquimatosas
6.
Tissue Cell ; 63: 101326, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32223953

RESUMEN

OBJECTIVES: We aim to develop a 3D-bilayer collagen (COL) membrane reinforced with nano beta-tricalcium-phosphate (nß-TCP) particles and to evaluate its bone regeneration in combination with leukocyte-platelet-rich fibrin (L-PRF) in vivo. BACKGROUND DATA: L-PRF has exhibited promising results as a cell carrier in bone regeneration in a number of clinical studies, however there are some studies that did not confirm the positive results of L-PRF application. METHODS: Mechanical & physiochemical characteristics of the COL/nß-TCP membrane (1/2 & 1/4) were tested. Proliferation and osteogenic differentiation of seeded cells on bilayer collagen/nß-TCP thick membrane was examined. Then, critical-sized calvarial defects in 8 white New Zealand rabbits were filled with either Col, Col/nß-TCP, Col/nß-TCP combined with L-PRF membrane, or left empty. New bone formation (NBF) was measured histomorphometrically 4 & 8 weeks postoperatively. RESULTS: Compressive modulus increases while porosity decreases with higher ß-TCP concentrations. Mechanical properties improve, with 89 % porosity (pore size ∼100 µm) in the bilayer-collagen/nß-TCP membrane. The bilayer design also enhances the proliferation and ALP activity. In vivo study shows no significant difference among test groups at 4 weeks, but Col/nß-TCP + L-PRF demonstrates more NBF compared to others (P < 0.05) after 8 weeks. CONCLUSION: The bilayer-collagen/nß-TCP thick membrane shows promising physiochemical in vitro results and significant NBF, as ¾ of the defect is filled with lamellar bone when combined with L-PRF membrane.


Asunto(s)
Enfermedades Óseas/terapia , Regeneración Ósea/genética , Colágeno/farmacología , Fibrina Rica en Plaquetas/metabolismo , Animales , Enfermedades Óseas/genética , Enfermedades Óseas/patología , Colágeno/química , Humanos , Leucocitos/metabolismo , Membranas/química , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Fibrina Rica en Plaquetas/química , Conejos
7.
Adv Exp Med Biol ; 1084: 17-43, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29423675

RESUMEN

Malignant bone tumors, although quite rare, are one of the causes of death in children and adolescents. Surgery as a common and primary treatment for removal of virtually bone cancer cause large bone defects. Thus, restoration of hard tissues like bone and cartilage after surgical tumor resection needs efficient therapeutic approaches. Tissue engineering (TE) is a powerful approach which has provided hope for restoration, maintenance, or improvement of damaged tissues. This strategy generally supplies a three-dimensional scaffold as an active substrate to support cell recruitment, infiltration, and proliferation for neo-tissues. The scaffold mimics the natural extracellular matrix (ECM) of tissue which needs to be regenerated. The use of potent cell sources such as mesenchymal stem cells (MSCs) has also led to remarkable progresses in hard tissue regeneration. Combination of living cells and various biomaterials have continuously evolved over the past decades to improve the process of regeneration. This chapter describes various strategies used in TE and highlights recent advances in cell-loaded constructs. We herein focus on cell-based scaffold approach utilized in hard tissue engineering and parameters determining a clinically efficient outcome. Also, we attempt to identify the potential as well as shortcomings of pre-loaded scaffolds for future therapeutic applications.


Asunto(s)
Neoplasias Óseas , Huesos , Células Madre Mesenquimatosas , Regeneración , Ingeniería de Tejidos , Neoplasias Óseas/cirugía , Neoplasias Óseas/terapia , Huesos/cirugía , Cartílago , Humanos , Andamios del Tejido
8.
J Mater Sci Mater Med ; 30(1): 1, 2018 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-30564959

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α), a well-studied angiogenesis pathway, plays an essential role in angiogenesis-osteogenesis coupling. Targeting the HIF-1a pathway frequently leads to successful reconstruction of large-sized bone defects through promotion of angiogenesis. Dimethyloxalylglycine (DMOG) small molecule regulates the stability of HIF-1α at normal oxygen tension by mimicking hypoxia, which subsequently accelerates angiogenesis. The current study aims to develop a novel construct by seeding adipose derived mesenchymal stem cells (ADMSCs) onto a scaffold that contains DMOG to induce angiogenesis and regeneration of a critical size calvarial defect in a rat model. The spongy scaffolds have been synthesized in the presence and absence of DMOG and analyzed in terms of morphology, porosity, pore size, mechanical properties and DMOG release profile. The effect of DMOG delivery on cellular behaviors of adhesion, viability, osteogenic differentiation, and angiogenesis were subsequently evaluated under in vitro conditions. Histological analysis of cell-scaffold constructs were also performed following transplantation into the calvarial defect. Physical characteristics of fabricated scaffolds confirmed higher mechanical strength and surface roughness of DMOG-loaded scaffolds. Scanning electron microscopy (SEM) images and MTT assay demonstrated the attachment and viability of ADMSCs in the presence of DMOG, respectively. Osteogenic activity of ADMSCs that included alkaline phosphatase (ALP) activity and calcium deposition significantly increased in the DMOG-loaded scaffold. Computed tomography (CT) imaging combined with histomorphometry and immunohistochemistry analysis showed enhanced bone formation and angiogenesis in the DMOG-loaded scaffolds. Therefore, spongy scaffolds that contained DMOG and had angiogenesis ability could be utilized to enhance bone regeneration of large-sized bone defects.


Asunto(s)
Ácido Algínico/química , Aminoácidos Dicarboxílicos/farmacología , Desarrollo Óseo , Fosfatos de Calcio/química , Gelatina/química , Andamios del Tejido , Aminoácidos Dicarboxílicos/administración & dosificación , Animales , Materiales Biocompatibles , Huesos/lesiones , Adhesión Celular/efectos de los fármacos , Supervivencia Celular , Liberación de Fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Células Madre Mesenquimatosas , Microscopía Electrónica de Rastreo , Neovascularización Fisiológica , Ratas , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
J Biomed Mater Res B Appl Biomater ; 105(7): 1767-1777, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-27186846

RESUMEN

We sought to assess the effects of coculturing mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs) in the repair of dog mandible bone defects. The cells were delivered in ß-tricalcium phosphate scaffolds coated with poly lactic co-glycolic acid microspheres that gradually release vascular endothelial growth factor (VEGF). The complete scaffold and five partial scaffolds were implanted in bilateral mandibular body defects in eight beagles. The scaffolds were examined histologically and morphometrically 8 weeks after implantation. Histologic staining of the decalcified scaffolds demonstrated that bone formation was greatest in the VEGF/MSC scaffold (63.42 ± 1.67), followed by the VEGF/MSC/EPC (47.8 ± 1.87) and MSC/EPC (45.21 ± 1.6) scaffolds, the MSC scaffold (34.59 ± 1.49), the VEGF scaffold (20.03 ± 1.29), and the untreated scaffold (7.24 ± 0.08). Hence, the rate of new bone regeneration was highest in scaffolds containing MSC, either mixed with EPC or incorporating VEGF. Adding both EPC and VEGF with the MSC was not necessary. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1767-1777, 2017.


Asunto(s)
Células Inmovilizadas , Células Progenitoras Endoteliales , Mandíbula , Células Madre Mesenquimatosas , Osteogénesis/efectos de los fármacos , Ingeniería de Tejidos , Andamios del Tejido/química , Animales , Células Inmovilizadas/citología , Células Inmovilizadas/trasplante , Técnicas de Cocultivo , Perros , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Mandíbula/citología , Mandíbula/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
Mater Sci Eng C Mater Biol Appl ; 69: 780-8, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27612772

RESUMEN

Bone tissue engineering is sought to apply strategies for bone defects healing without limitations and short-comings of using either bone autografts or allografts and xenografts. The aim of this study was to fabricate a thin layer poly(lactic-co-glycolic) acid (PLGA) coated beta-tricalcium phosphate (ß-TCP) scaffold with sustained release of vascular endothelial growth factor (VEGF). PLGA coating increased compressive strength of the ß-TCP scaffolds significantly. For in vitro evaluations, canine mesenchymal stem cells (cMSCs) and canine endothelial progenitor cells (cEPCs) were isolated and characterized. Cell proliferation and attachment were demonstrated and the rate of cells proliferation on the VEGF released scaffold was significantly more than compared to the scaffolds with no VEGF loading. A significant increase in expression of COL1 and RUNX2 was indicated in the scaffolds loaded with VEGF and MSCs compared to the other groups. Consequently, PLGA coated ß-TCP scaffold with sustained and localized release of VEGF showed favourable results for bone regeneration in vitro, and this scaffold has the potential to use as a drug delivery device in the future.


Asunto(s)
Huesos/fisiología , Fosfatos de Calcio/química , Materiales Biocompatibles Revestidos/química , Ácido Láctico/química , Ácido Poliglicólico/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Huesos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Fuerza Compresiva , Perros , Liberación de Fármacos , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/ultraestructura , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/ultraestructura , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Porosidad , Reacción en Cadena en Tiempo Real de la Polimerasa , Difracción de Rayos X
11.
J Oral Implantol ; 39(4): 433-43, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23964777

RESUMEN

This study aimed to investigate effects of dental pulp stem cells (DPSCs) on regeneration of a defect experimentally created in the periodontium of a canine model. Surgically created mesial 3-walled periodontal defects with ligature-induced periodontitis were produced bilaterally in the first lower premolar teeth of 10 mongrel dogs. Simultaneously, DPSCs were derived from the maxillary premolar teeth of the same dogs. Four weeks after creation of the periodontitis model, autologous passaged-3 DPSCs combined with Bio-Oss were implanted on one side as the test group. On the other side, only Bio-Oss was implanted as a control. Eight weeks after surgery, regeneration of the periodontal defects was evaluated histologically and histomorphometrically in terms of bone, periodontal ligament (PDL), and cement formation. Histologically, in all test specimens (10 defects), regeneration of cementum, bone, and PDL was observed. In the control groups, although we observed the regeneration of bone in all defects, the formation of cementum was seen in 9 defects and PDL was seen in 8 defects. Histomorphometric analyses showed that the amount of regenerated cementum and PDL in the test groups (3.83 ± 1.32 mm and 3.30 ± 1.12 mm, respectively) was significantly higher than that of the control groups (2.42 ± 1.40 mm and 1.77 ± 1.27 mm, respectively; P < .05). A biocomplex consisting of DPSCs and Bio-Oss would be promising in regeneration of periodontal tissues.


Asunto(s)
Pérdida de Hueso Alveolar/cirugía , Regeneración Ósea , Pulpa Dental/citología , Periodontitis/cirugía , Trasplante de Células Madre , Adipogénesis , Animales , Sustitutos de Huesos , Células Cultivadas , Condrogénesis , Cemento Dental/fisiología , Perros , Masculino , Minerales , Ligamento Periodontal/fisiología , Regeneración , Andamios del Tejido
12.
Arch Iran Med ; 15(7): 422-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22724879

RESUMEN

BACKGROUND: Osteoarthritis (OA) is a progressive disorder of the joints caused by gradual loss of articular cartilage, which naturally possesses a limited regenerative capacity. In the present study, the potential of intra-articular injection of mesenchymal stem cells (MSCs) has been evaluated in six osteoarthritic patients. METHODS: Six female volunteers, average age of 54.56 years, with radiologic evidence of knee OA that required joint replacement surgery were selected for this study. About 50 ml bone marrow was aspirated from each patient and taken to the cell laboratory, where MSCs were isolated and characterized in terms of some surface markers. About 20-24 × 10(6) passaged-2 cells were prepared and tested for microbial contamination prior to intra-articular injection. RESULTS: During a one-year follow-up period, we found no local or systemic adverse events. All patients were partly satisfied with the results of the study. Pain, functional status of the knee, and walking distance tended to be improved up to six months post-injection, after which pain appeared to be slightly increased and patients' walking abilities slightly decreased. Comparison of magnetic resonance images (MRI) at baseline and six months post-stem cell injection displayed an increase in cartilage thickness, extension of the repair tissue over the subchondral bone and a considerable decrease in the size of edematous subchondral patches in three out of six patients. CONCLUSION: The results indicated satisfactory effects of intra-articular injection of MSCs in patients with knee OA.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Osteoartritis de la Rodilla/terapia , Adulto , Femenino , Estudios de Seguimiento , Humanos , Inyecciones Intraarticulares , Imagen por Resonancia Magnética , Persona de Mediana Edad , Osteoartritis de la Rodilla/patología , Trasplante Autólogo
13.
Int J Fertil Steril ; 6(3): 147-56, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24520432

RESUMEN

Advances in stem cell biotechnology hold great promise in the field of tissue engineering and regenerative medicine. Of interest are marrow mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs). In addition, amniotic fluid stem cells (AFSCs) have attracted attention as a viable choice following the search for an alternative stem cell source. Investigators are interested in these cells because they come from the amniotic fluid that is routinely discarded after birth. There have been multiple investigations conducted worldwide in an attempt to better understand AF-SCs in terms of their potential use in regenerative medicine. In this review we give a brief introduction of amniotic fluid followed by a description of the cells present within this fluid. Their history related to stem cell discovery in the amniotic fluid as well as the main characteristics of AF-SCs are discussed. Finally, we elaborate on the potential for these cells to promote regeneration of various tissue defects, including fetal tissue, the nervous system, heart, lungs, kidneys, bones, and cartilage.

14.
Arch Iran Med ; 14(2): 96-103, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21361715

RESUMEN

BACKGROUND: Despite the suitability of a mouse model for preclinical investigations; little is known regarding mesenchymal stem cells derived from murine amniotic fluid. This is the subject of the present study.  METHODS:   Amniotic fluid was collected from NMRI mice during the second weeks of pregnancy and plated. The cells that adhered to the culture surfaces were propagated with three successive subcultures and then characterized. To determine the differentiation potential, the cells were cultivated under osteogenic, adipogenic, and chondrogenic conditions, and followed by specific staining and RT-PCR analysis for differentiation. The proliferative potential of the cells were measured with clonogenic assays, population doubling time and number and by growth curve plotting. Cellular aging was investigated with the senescence-associated ß-galactosidase staining method. RESULTS: The amniotic fluid primary cell culture was composed of round flattened and fibroblastic cells. The latter dominated the culture after several passages. Successful tripotent differentiation of the isolated cells into bone, cartilage and adipose cells were indicative of their mesenchymal stem cells nature. The isolated cells appeared to be relatively proliferative cells as confirmed by the population doubling time value which was equal to about 69 hours. Furthermore, the cells were relatively clonogenic and they tended to initiate proliferation immediately after plating (there was no lag phase in their growth curve). ß-galactosidase positive cells were first observed at passage 3 and increased in number with subsequent passages. CONCLUSIONS: Collectively it was concluded that murine amniotic fluid contained mesenchymal stem cells with relatively high proliferation property and typical tripotent differentiation potential.  


Asunto(s)
Líquido Amniótico/citología , Diferenciación Celular , Proliferación Celular , Senescencia Celular , Células Madre Mesenquimatosas/citología , Animales , Células Cultivadas , Femenino , Citometría de Flujo , Receptores de Hialuranos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Modelos Animales , Modelos Biológicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Antígenos Thy-1/metabolismo , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA