Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Small ; 12(10): 1342-50, 2016 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-26756453

RESUMEN

Assessing mesenchymal stem cell (MSC) differentiation status is crucial to verify therapeutic efficacy and optimize treatment procedures. Currently, this involves destructive methods including antibody-based protein detection and polymerase chain reaction gene analysis, or laborious and technically challenging genetic reporters. Development of noninvasive methods for real-time differentiation status assessment can greatly benefit MSC-based therapies. This report introduces a nanoparticle-based sensing platform that encapsulates two molecular beacon (MB) probes within the same biodegradable polymeric nanoparticles. One MB targets housekeeping gene glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as an internal reference, while another detects alkaline phosphatase (ALP), a functional biomarker. Following internalization, MBs are gradually released as the nanoparticle degrades. GAPDH MBs provide a stable reference signal throughout the monitoring period (18 days) regardless of differentiation induction. Meanwhile, ALP mRNA undergoes well-defined dynamics with peak expression observed during early stages of osteogenic differentiation. By normalizing ALP-MB signal with GAPDH-MB, changes in ALP expression can be monitored, to noninvasively validate osteogenic differentiation. As proof-of-concept, a dual-colored nanosensor is applied to validate MSC osteogenesis on 2D culture and polycaprolactone films containing osteo-inductive tricalcium phospate.


Asunto(s)
Técnicas Biosensibles/instrumentación , Diferenciación Celular , Células Madre Mesenquimatosas/citología , Nanotecnología/instrumentación , Osteogénesis , Fosfatos de Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Poliésteres/farmacología
2.
Mol Pharm ; 11(7): 2126-33, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-24779855

RESUMEN

In spite of recognized limitations in capturing species-specific responses and high costs, rodent models remain commonly used in prostate cancer metastasis research, due largely to the lack of available alternatives. We aim to develop an in vitro culture system to study prostate cancer response to a simulated bone microenvironment, which may be used to understand early events in prostate metastasis to bone or for drug screening applications. To achieve this, mesenchymal stem cells and endothelial cells were isolated and cocultured to form a vascularized bone analogue. Endothelial cells were found to exert osteopotentiating effects on mesenchymal stem cell differentiation, and reciprocal effects by the stromal cells were found to stimulate vasculogenic responses, suggesting the possible utility of this system to elicit three-way interactions between endothelial, mesenchymal, and prostate cancer cells. We further developed the use of fluorescently labeled cells which could be used to concurrently track cellular migration, proliferation, and morphometric analysis. We demonstrate the concurrent, real-time visualization of prostate cancer and endothelial cells, which may be useful for evaluation of spatiotemporal changes at a single-cell level. When prostate cancer cell proliferation on various substrates was measured, it was found that the use of coculture systems may provide a better reflection of conditions in vivo, highlighting the potential utility as a model system.


Asunto(s)
Células Endoteliales/patología , Células Madre Mesenquimatosas/patología , Neoplasias de la Próstata/patología , Células de la Médula Ósea/patología , Huesos/patología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo/métodos , Humanos , Masculino
3.
Stem Cells ; 30(9): 1911-24, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22761003

RESUMEN

Umbilical cord blood-derived endothelial colony-forming cells (UCB-ECFC) show utility in neovascularization, but their contribution to osteogenesis has not been defined. Cocultures of UCB-ECFC with human fetal-mesenchymal stem cells (hfMSC) resulted in earlier induction of alkaline phosphatase (ALP) (Day 7 vs. 10) and increased mineralization (1.9×; p < .001) compared to hfMSC monocultures. This effect was mediated through soluble factors in ECFC-conditioned media, leading to 1.8-2.2× higher ALP levels and a 1.4-1.5× increase in calcium deposition (p < .01) in a dose-dependent manner. Transcriptomic and protein array studies demonstrated high basal levels of osteogenic (BMPs and TGF-ßs) and angiogenic (VEGF and angiopoietins) regulators. Comparison of defined UCB and adult peripheral blood ECFC showed higher osteogenic and angiogenic gene expression in UCB-ECFC. Subcutaneous implantation of UCB-ECFC with hfMSC in immunodeficient mice resulted in the formation of chimeric human vessels, with a 2.2-fold increase in host neovascularization compared to hfMSC-only implants (p = .001). We conclude that this study shows that UCB-ECFC have potential in therapeutic angiogenesis and osteogenic applications in conjunction with MSC. We speculate that UCB-ECFC play an important role in skeletal and vascular development during perinatal development but less so in later life when expression of key osteogenesis and angiogenesis genes in ECFC is lower.


Asunto(s)
Endotelio Vascular/citología , Sangre Fetal/citología , Células Madre Mesenquimatosas/citología , Osteogénesis/fisiología , Animales , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Sangre Fetal/metabolismo , Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis por Micromatrices
4.
Stem Cells ; 27(1): 126-37, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18832592

RESUMEN

Mesenchymal stem cells (MSCs) from human adult bone marrow (haMSCs) represent a promising source for bone tissue engineering. However, their low frequencies and limited proliferation restrict their clinical utility. Alternative postnatal, perinatal, and fetal sources of MSCs appear to have different osteogenic capacities, but have not been systematically compared with haMSCs. We investigated the proliferative and osteogenic potential of MSCs from human fetal bone marrow (hfMSCs), human umbilical cord (hUCMSCs), and human adult adipose tissue (hATMSCs), and haMSCs, both in monolayer cultures and after loading into three-dimensional polycaprolactone-tricalcium-phosphate scaffolds.Although all MSCs had comparable immunophenotypes, only hfMSCs and hUCMSCs were positive for the embryonic pluripotency markers Oct-4 and Nanog. hfMSCs expressed the lowest HLA-I level (55% versus 95%-99%) and the highest Stro-1 level (51% versus 10%-27%), and had the greatest colony-forming unit-fibroblast capacity (1.6x-2.0x; p < .01) and fastest doubling time (32 versus 54-111 hours; p < .01). hfMSCs had the greatest osteogenic capacity, as assessed by von-Kossa staining, alkaline phosphatase activity (5.1x-12.4x; p < .01), calcium deposition (1.6x-2.7x in monolayer and 1.6x-5.0x in scaffold culture; p < .01), calcium visualized on micro-computed tomography (3.9x17.6x; p < .01) and scanning electron microscopy, and osteogenic gene induction. Two months after implantation of cellular scaffolds in immunodeficient mice, hfMSCs resulted in the most robust mineralization (1.8x-13.3x; p < .01).The ontological and anatomical origins of MSCs have profound influences on the proliferative and osteogenic capacity of MSCs. hfMSCs had the most proliferative and osteogenic capacity of the MSC sources, as well as being the least immunogenic, suggesting they are superior candidates for bone tissue engineering.


Asunto(s)
Células Madre Adultas/citología , Huesos/fisiología , Feto/citología , Células Madre Mesenquimatosas/citología , Osteogénesis , Ingeniería de Tejidos , Tejido Adiposo/citología , Adulto , Células Madre Adultas/efectos de los fármacos , Animales , Huesos/efectos de los fármacos , Fosfatos de Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Implantes Experimentales , Lactante , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/ultraestructura , Ratones , Ratones SCID , Persona de Mediana Edad , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Poliésteres/farmacología , Andamios del Tejido , Cordón Umbilical/citología
5.
Stem Cells ; 27(8): 1921-31, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19544438

RESUMEN

Stem cell transplantation for regenerative medicine has made significant progress in various injury models, with the development of modalities to track stem cell fate and migration post-transplantation being currently pursued rigorously. Magnetic resonance imaging (MRI) allows serial high-resolution in vivo detection of transplanted stem cells labeled with iron oxide particles, but has been hampered by low labeling efficiencies. Here, we describe the use of microgel iron oxide (MGIO) particles of diameters spanning 100-750 nm for labeling human fetal mesenchymal stem cells (hfMSCs) for MRI tracking. We found that MGIO particle uptake by hfMSCs was size dependent, with 600-nm MGIO (M600) particles demonstrating three- to sixfold higher iron loading than the clinical particle ferucarbotran (33-263 versus 9.6-42.0 pg iron/hfMSC; p < .001). Cell labeling with either M600 particles or ferucarbotran did not affect either cellular proliferation or tri-lineage differentiation into osteoblasts, adipocytes, and chondrocytes, despite differences in gene expression on a genome-wide microarray analysis. Cell tracking in a rat photothrombotic stroke model using a clinical 1.5-T MRI scanner demonstrated the migration of labeled hfMSCs from the contralateral cortex to the stroke injury, with M600 particles achieving a five- to sevenfold higher sensitivity for MRI detection than ferucarbotran (p < .05). However, model-related cellular necrosis and acute inflammation limited the survival of hfMSCs beyond 5-12 days. The use of M600 particles allowed high detection sensitivity with low cellular toxicity to be achieved through a simple incubation protocol, and may thus be useful for cellular tracking using standard clinical MRI scanners.


Asunto(s)
Compuestos Férricos/química , Células Madre Fetales/química , Imagen por Resonancia Magnética/métodos , Células Madre Mesenquimatosas/química , Nanopartículas/química , Animales , Medios de Contraste/metabolismo , Femenino , Células Madre Fetales/citología , Humanos , Células Madre Mesenquimatosas/citología , Embarazo , Ratas , Ratas Wistar
6.
J Biomed Mater Res A ; 102(7): 2197-207, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23907895

RESUMEN

Geometric cues have been used for a variety of cell regulation and tissue regenerative applications. While the function of geometric cues is being recognized, their stability and degradation behaviors are not well known. Here, we studied the influence of degradation on uniaxial-stretch-induced poly(ε-caprolactone) (UX-PCL) ridge/groove arrays and further cellular responses. Results from accelerated hydrolysis in vitro showed that UX-PCL ridge/groove arrays followed a surface-controlled erosion, with an overall geometry remained even at ∼45% film weight loss. Compared to unstretched PCL flat surfaces and/or ridge/groove arrays, UX-PCL ridge/groove arrays achieved an enhanced morphological stability against degradation. Over the degradation period, UX-PCL ridge/groove arrays exhibited an "S-shape" behavior of film weight loss, and retained more stable surface hydrophilicity and higher film mechanical properties than those of unstretched PCL surfaces. Human mesenchymal stem cells (MSCs) aligned better toward UX-PCL ridge/groove arrays when the geometries were remained intact, and became sensitive with gradually declined nucleus alignment and elongation to the geometric degradation of ridges. We speculate that uniaxial stretching confers UX-PCL ridge/groove arrays with enhanced stability against degradation in erosive environment. This study provides insights of how degradation influences geometric cues and further cell responses, and has implications for the design of biomaterials with stability-enhanced geometric cues for long-term tissue regeneration.


Asunto(s)
Biomimética , Células Madre Mesenquimatosas/metabolismo , Poliésteres/metabolismo , Ensayo de Materiales , Microscopía Electrónica de Rastreo , Poliésteres/química , Propiedades de Superficie
7.
Tissue Eng Part A ; 19(7-8): 893-904, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23102089

RESUMEN

Clinical translation of bone tissue engineering approaches for fracture repair has been hampered by inadequate vascularization required for maintaining cell survival, skeletal regeneration, and remodeling. The potential of vasculature formation within tissue-engineered grafts depends on various factors, including an appropriate choice of scaffold and its microarchitectural design for the support of tissue ingrowth and vessel infiltration, vasculogenic potential of cell types and mechanostimulation on cells to enhance cytokine expression. Here, we demonstrated the effect of biomechanical stimulation on vasculogenic and bone-forming capacity of umbilical-cord-blood endothelial progenitor cells (UCB-EPC) and human fetal bone marrow-derived mesenchymal stem cell (hfMSC) seeded within macroporous scaffolds and cocultured dynamically in a biaxial bioreactor. Dynamically cultured EPC/hfMSC constructs generated greater mineralization and calcium deposition consistently over 14 days of culture (1.7-fold on day 14; p<0.05). However, in vitro vessel formation was not observed as compared to an extensive EPC-vessel network formed under static culture on day 7. Subsequent subcutaneous implantations in NOD/SCID mice showed 1.4-fold higher human:mouse cell chimerism (p<0.001), with a more even cellular distribution throughout the dynamically cultured scaffolds. In addition, there was earlier evidence of vessel infiltration into the scaffold and a trend toward increased ectopic bone formation, suggesting improved efficacy and cellular survival through early vascularization upon biomechanical stimulation. The integrative use of bioreactor culture systems with macroporous scaffolds and cocultured osteogenic and vasculogenic cells promotes maturation of EPC/hfMSC-scaffold grafts necessary for vascularized bone tissue engineering applications.


Asunto(s)
Reactores Biológicos , Huesos/fisiología , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Células Madre/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Huesos/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Fosfatos de Calcio/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Feto/citología , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones SCID , Neovascularización Fisiológica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Poliésteres/farmacología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Cordón Umbilical/citología
8.
Macromol Biosci ; 13(6): 799-807, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23606448

RESUMEN

Human mesenchymal stem cells (MSCs) derived from various origins show varied differentiation capability. Recent work shows that cell shape manipulation via micropatterning can modulate the differentiation of bone-marrow-derived MSCs. Herein, the effect of micropatterning on the myogenesis of MSCs isolated from three different sources (bone marrow, fetal tissue, and adipose) is reported. All the well-aligned cells, regardless of source, predominantly commit to myogenic lineage, as shown by the significant upregulation of myogenic gene markers and positive myosin heavy chain staining. It is demonstrated that our novel micropattern can be used as a generic platform for inducing myogenesis of MSCs from different sources and may also have the potential to be extended to induce other lineage commitment.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Linaje de la Célula , Células Madre Mesenquimatosas/citología , Desarrollo de Músculos , Tejido Adiposo/citología , Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Dimetilpolisiloxanos/farmacología , Feto/citología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/metabolismo , Microscopía Fluorescente , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/genética , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Stem Cells Transl Med ; 2(11): 839-47, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24106336

RESUMEN

The term placenta is a highly vascularized tissue and is usually discarded upon birth. Our objective was to isolate clinically relevant quantities of fetal endothelial colony-forming cells (ECFCs) from human term placenta and to compare them to the well-established donor-matched umbilical cord blood (UCB)-derived ECFCs. A sorting strategy was devised to enrich for CD45-CD34+CD31Lo cells prior to primary plating to obtain pure placental ECFCs (PL-ECFCs) upon culture. UCB-ECFCs were derived using a well-described assay. PL-ECFCs were fetal in origin and expressed the same cell surface markers as UCB-ECFCs. Most importantly, a single term placenta could yield as many ECFCs as 27 UCB donors. PL-ECFCs and UCB-ECFCs had similar in vitro and in vivo vessel forming capacities and restored mouse hind limb ischemia in similar proportions. Gene expression profiles were only minimally divergent between PL-ECFCs and UCB-ECFCs, probably reflecting a vascular source versus a circulating source. Finally, PL-ECFCs and UCB-ECFCs displayed similar hierarchies between high and low proliferative colonies. We report a robust strategy to isolate ECFCs from human term placentas based on their cell surface expression. This yielded much larger quantities of ECFCs than UCB, but the cells were comparable in immunophenotype, gene expression, and in vivo functional ability. We conclude that PL-ECFCs have significant bio-banking and clinical translatability potential.


Asunto(s)
Antígenos de Superficie/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Feto/citología , Feto/metabolismo , Placenta/citología , Placenta/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Superficie/genética , Biomarcadores/metabolismo , Procesos de Crecimiento Celular/fisiología , Femenino , Sangre Fetal/citología , Sangre Fetal/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Embarazo , Estudios Prospectivos , Transcriptoma
10.
Methods Mol Biol ; 614: 135-47, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20225041

RESUMEN

Human fetal mesenchymal stem cells (hfMSC) demonstrate extensive expansion and differentiation capacities and are hence being studied for use in stem cell therapeutics, including gene delivery. With advanced prenatal diagnosis, fetal gene therapy represents an additional avenue for the treatment of inherited deficiencies. We have recently demonstrated harvesting of first-trimester fMSC from fetal blood for ex vivo genetic engineering to introduce genes of interest, and finally intra-uterine transplantation (IUT) of these cells to the fetus. Here we discuss methods in the harvesting of hfMSC, lentiviral transduction to introduce genes of interest, and in vitro methods to characterise transgene expression.


Asunto(s)
Feto/citología , Vectores Genéticos , Lentivirus/genética , Células Madre Mesenquimatosas/citología , Transducción Genética , Línea Celular , Humanos , Microscopía Fluorescente
11.
Biomaterials ; 31(4): 608-20, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19836073

RESUMEN

Tissue-engineered bone grafts (TEBG) require highly osteogenic cell sources for use in fracture repair applications. Compared to other sources of mesenchymal stem cells (MSC), human fetal MSC (hfMSC) have recently been shown to be more proliferative and osteogenic. We studied the functional performance of hfMSC-mediated TEBG in 7 mm rat femoral critical-sized bone defects (CSD). Dynamically-cultured and osteogenically-primed hfMSC seeded onto macroporous poly-epsilon-caprolactone tri-calcium phosphate scaffolds were transplanted into CSDs. After 12 weeks, hfMSC-mediated TEBG induced 2.1x more new bone formation (43.3+/-10.5 vs. 21.0+/-7.4 mm(3), p<0.05), with greater compact and woven bone, and a 9.8x increase in stiffness (3.9+/-1.7 vs. 0.4+/-0.3 mNm/degree, p<0.05) compared to acellular scaffolds, such that only animals transplanted with TEBG underwent full fracture repair of the CSD. Although hfMSC survived for <4 weeks, by 4 weeks they were associated with a 3.9x larger vasculature network in the defect area (35.2+/-11.1 vs. 6.5+/-3.6 mm(3)p<0.05), suggesting an important role for hfMSC in the promotion of neo-vasculogenesis. We speculate that hfMSC-mediated healing of the CSD by stimulating neo-vascularization through as yet undetermined mechanisms. This proof-of-principle study demonstrates the utility of primitive MSC for bone regeneration, and may be of relevance to vascularization in other areas of regenerative medicine.


Asunto(s)
Fémur/irrigación sanguínea , Feto/citología , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica/fisiología , Osteogénesis/fisiología , Ingeniería de Tejidos/métodos , Animales , Humanos , Inmunohistoquímica , Masculino , Células Madre Mesenquimatosas/fisiología , Microscopía de Fuerza Atómica , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Tomografía Computarizada por Rayos X
12.
Biomaterials ; 31(12): 3296-306, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20116846

RESUMEN

In vivo tracking of stem cells after transplantation is crucial for understanding cell-fate and therapeutic efficacy. By labelling stem cells with magnetic particles, they can be tracked by Magnetic Resonance Imaging (MRI). We previously demonstrated that microgel iron oxide nanoparticle (MGIO) provide superior tracking sensitivity over commercially available particles. Here, we describe the synthesis of MGIO and report on their morphology, hydrodynamic diameters (87-766 nm), iron oxide weight content (up to 82%) and magnetization characteristics (M(s)=52.9 Am(2)/kg, M(R)=0.061 Am(2)/kg and H(c)=0.672 A/m). Their MR relaxation characteristics are comparable to those of theoretical models and represent the first such correlation between model and real particles of varying diameters. A labelling study of primary endothelial progenitor cells also confirms that MGIO is an efficient label regardless of cell type. The facile synthesis of MGIO makes it a useful tool for the studying of relaxation induced by magnetic particles and cellular tracking by MRI.


Asunto(s)
Endotelio/citología , Compuestos Férricos/química , Geles , Imagen por Resonancia Magnética , Nanopartículas , Células Madre/citología , Microscopía Electrónica de Transmisión
13.
Biomaterials ; 30(12): 2241-51, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19200592

RESUMEN

Cell-sheet assemblies are currently being studied for tissue engineering. However, tissues engineered from completely biological cell sheets lack substrate cues and possess poor mechanical strength. Recent studies demonstrate the use of synthetic bioresorbable films as scaffolds that may address these issues. Here, we describe the application of a micro-thin, biaxially-stretched polycaprolactone (muXPCL) with surface modifications for layered tissue engineering, and present the results of biphasic cell-sheet constructs using surfaces optimised for specific cell types. Polyacrylic acid (PAAc) was grafted onto muXPCL film surfaces by low-pressure plasma immobilisation. This provided a surface suitable for perivascular cells, forming the medial compartment. Subsequently, endothelial progenitor cell (EPC)-selective CD34 antibody was conjugated onto the reverse surface (intimal compartment) to select and anchor EPCs for improved adhesion and proliferation. Using the blood vessel as a model, a biphasic culture system was then setup to represent a tunica intima (endothelial cells) and tunica media (smooth muscle cells). When suitable cell types were cultured in the corresponding compartments, confluent layers of the respective populations were achieved distinctively from each other. These results demonstrate the use of muXPCL films with cell-selective modifications for layered co-cultures towards the generation of stratified tissue.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Madre/citología , Ingeniería de Tejidos/métodos , Cordón Umbilical/irrigación sanguínea , Cordón Umbilical/citología , Resinas Acrílicas/química , Antígenos CD34/metabolismo , Separación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Humanos , Células Madre/metabolismo , Propiedades de Superficie , Resistencia a la Tracción
14.
IEEE Trans Biomed Eng ; 56(9): 2331-4, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19497809

RESUMEN

Hematopoietic stem cells (HSCs) from the human placenta and umbilical cord blood (UCB) provide a rich source of highly proliferative cells for many clinical uses with advantages over traditional sources like the bone marrow and periphery blood. However, the key current constraint with this source of HSCs is the inadequate number of HSCs cells that can be harvested in a single collection using current approaches, which render a large number of collections unusable on their own, even for pediatric patients. This paper will present the development of a device to enable more efficient harvesting of HSCs from placentas, which can be used ex utero, upon the discharge of placentas after deliveries. The device can be used to facilitate a two-fraction collection process. Results, in terms of mononucleated cells (MNCs) count, CD34+ cells count, as well as flow cytometry, will be furnished to verify the effectiveness of the developed system.


Asunto(s)
Automatización/instrumentación , Separación Celular/instrumentación , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Placenta/citología , Antígenos CD , Técnicas de Cultivo de Célula , Diseño de Equipo , Femenino , Citometría de Flujo , Humanos , Leucocitos Mononucleares , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA