Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transl Vis Sci Technol ; 5(4): 6, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27486556

RESUMEN

PURPOSE: We assessed the long-term efficacy and safety of human retinal progenitor cells (hRPC) using established rodent models. METHODS: Efficacy of hRPC was tested initially in Royal College of Surgeons (RCS) dystrophic rats immunosuppressed with cyclosporine/dexamethasone. Due to adverse effects of dexamethasone, this drug was omitted from a subsequent dose-ranging study, where different hRPC doses were tested for their ability to preserve visual function (measured by optokinetic head tracking) and retinal structure in RCS rats at 3 to 6 months after grafting. Safety of hRPC was assessed by subretinal transplantation into wild type (WT) rats and NIH-III nude mice, with analysis at 3 to 6 and 9 months after grafting, respectively. RESULTS: The optimal dose of hRPC for preserving visual function/retinal structure in dystrophic rats was 50,000 to 100,000 cells. Human retinal progenitor cells integrated/survived in dystrophic and WT rat retina up to 6 months after grafting and expressed nestin, vimentin, GFAP, and ßIII tubulin. Vision and retinal structure remained normal in WT rats injected with hRPC and there was no evidence of tumors. A comparison between dexamethasone-treated and untreated dystrophic rats at 3 months after grafting revealed an unexpected reduction in the baseline visual acuity of dexamethasone-treated animals. CONCLUSIONS: Human retinal progenitor cells appear safe and efficacious in the preclinical models used here. TRANSLATIONAL RELEVANCE: Human retinal progenitor cells could be deployed during early stages of retinal degeneration or in regions of intact retina, without adverse effects on visual function. The ability of dexamethasone to reduce baseline visual acuity in RCS dystrophic rats has important implications for the interpretation of preclinical and clinical cell transplant studies.

2.
Genom Data ; 3: 70-4, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26484151

RESUMEN

Multipotent mesenchymal stromal cells derived from human placenta (pMSCs), and unrestricted somatic stem cells (USSCs) derived from cord blood share many properties with human bone marrow-derived mesenchymal stromal cells (bmMSCs) and are currently in clinical trials for a wide range of clinical settings. Here we present gene expression profiles of human cord blood-derived unrestricted somatic stem cells (USSCs), human placental-derived mesenchymal stem cells (hpMSCs), and human bone marrow-derived mesenchymal stromal cells (bmMSCs), all derived from four different donors. The microarray data are available on the ArrayExpress database (www.ebi.ac.uk/arrayexpress) under accession number E-TABM-880. Additionally, the data has been integrated into a public portal, www.stemformatics.org. Our data provide a resource for understanding the differences in MSCs derived from different tissues.

3.
J Clin Med ; 2(3): 115-35, 2013 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-26237066

RESUMEN

Haematopoietic stem cell (HSC) transplantation is an established cell-based therapy for a number of haematological diseases. To enhance this therapy, there is considerable interest in expanding HSCs in artificial niches prior to transplantation. This study compared murine HSC expansion supported through co-culture on monolayers of either undifferentiated mesenchymal stromal cells (MSCs) or osteoblasts. Sorted Lineage(-) Sca-1(+) c-kit(+) (LSK) haematopoietic stem/progenitor cells (HPC) demonstrated proliferative capacity on both stromal monolayers with the greatest expansion of LSK shown in cultures supported by osteoblast monolayers. After transplantation, both types of bulk-expanded cultures were capable of engrafting and repopulating lethally irradiated primary and secondary murine recipients. LSKs co-cultured on MSCs showed comparable, but not superior, reconstitution ability to that of freshly isolated LSKs. Surprisingly, however, osteoblast co-cultured LSKs showed significantly poorer haematopoietic reconstitution compared to LSKs co-cultured on MSCs, likely due to a delay in short-term reconstitution. We demonstrated that stromal monolayers can be used to maintain, but not expand, functional HSCs without a need for additional haematopoietic growth factors. We also demonstrated that despite apparently superior in vitro performance, co-injection of bulk cultures of osteoblasts and LSKs in vivo was detrimental to recipient survival and should be avoided in translation to clinical practice.

4.
Tissue Eng Part C Methods ; 18(5): 319-28, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22082070

RESUMEN

Hematopoietic stem cell (HSC) transplant is a well established curative therapy for some hematological malignancies. However, achieving adequate supply of HSC from some donor tissues can limit both its application and ultimate efficacy. The theory that this limitation could be overcome by expanding the HSC population before transplantation has motivated numerous laboratories to develop ex vivo expansion processes. Pioneering work in this field utilized stromal cells as support cells in cocultures with HSC to mimic the HSC niche. We hypothesized that through translation of this classic coculture system to a three-dimensional (3D) structure we could better replicate the niche environment and in turn enhance HSC expansion. Herein we describe a novel high-throughput 3D coculture system where murine-derived HSC can be cocultured with mesenchymal stem/stromal cells (MSC) in 3D microaggregates--which we term "micromarrows." Micromarrows were formed using surface modified microwells and their ability to support HSC expansion was compared to classic two-dimensional (2D) cocultures. While both 2D and 3D systems provide only a modest total cell expansion in the minimally supplemented medium, the micromarrow system supported the expansion of approximately twice as many HSC candidates as the 2D controls. Histology revealed that at day 7, the majority of bound hematopoietic cells reside in the outer layers of the aggregate. Quantitative polymerase chain reaction demonstrates that MSC maintained in 3D aggregates express significantly higher levels of key hematopoietic niche factors relative to their 2D equivalents. Thus, we propose that the micromarrow platform represents a promising first step toward a high-throughput HSC 3D coculture system that may enable in vitro HSC niche recapitulation and subsequent extensive in vitro HSC self-renewal.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Ingeniería de Tejidos/métodos , Animales , Agregación Celular/fisiología , Proliferación Celular , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
5.
Stem Cell Res ; 8(1): 58-73, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22099021

RESUMEN

Cells resembling bone marrow mesenchymal stem cells (MSC) have been isolated from many organs but their functional relationships have not been thoroughly examined. Here we compared the immunophenotype, gene expression, multipotency and immunosuppressive potential of MSC-like colony-forming cells from adult murine bone marrow (bmMSC), kidney (kCFU-F) and heart (cCFU-F), cultured under uniform conditions. All populations showed classic MSC morphology and in vitro mesodermal multipotency. Of the two solid organ-specific CFU-F, only kCFU-F displayed suppression of T-cell alloreactivity in vitro, albeit to a lesser extent than bmMSC. Quantitative immunophenotyping using 81 phycoerythrin-conjugated CD antibodies demonstrated that all populations contained high percentages of cells expressing diagnostic MSC surface markers (Sca1, CD90.2, CD29, CD44), as well as others noted previously on murine MSC (CD24, CD49e, CD51, CD80, CD81, CD105). Illumina microarray expression profiling and bioinformatic analysis indicated a correlation of gene expression of 0.88-0.92 between pairwise comparisons. All populations expressed approximately 66% of genes in the pluripotency network (Plurinet), presumably reflecting their stem-like character. Furthermore, all populations expressed genes involved in immunomodulation, homing and tissue repair, suggesting these as conserved functions for MSC-like cells in solid organs. Despite this molecular congruence, strong biases in gene and protein expression and pathway activity were seen, suggesting organ-specific functions. Hence, tissue-derived MSC may also retain unique properties potentially rendering them more appropriate as cellular therapeutic agents for their organ of origin.


Asunto(s)
Células de la Médula Ósea/citología , Inmunofenotipificación/métodos , Riñón/citología , Células Madre Mesenquimatosas/citología , Miocardio/citología , Transcriptoma/genética , Animales , Células de la Médula Ósea/metabolismo , Forma de la Célula , Ensayo de Unidades Formadoras de Colonias , Epítopos/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes/genética , Terapia de Inmunosupresión , Células Madre Mesenquimatosas/metabolismo , Ratones , Especificidad de Órganos/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
6.
Tissue Eng Part A ; 17(17-18): 2279-89, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21542698

RESUMEN

Tissue engineering approaches are currently being investigated for the restoration of myocardial function in heart failure patients, most commonly by combining cells with a substrate to form myocardial-like constructs (MCs). The final properties of these constructs are dependant on the characteristics of both the substrate and the cells used for fabrication. To create a construct with the appropriate mechanical properties required for any future therapeutic, we tailored an acrylated polypropylene glycol triol (aPPGT) substrate to the elastic modulus of heart tissue and then investigated the fabrication of MCs. We first assessed the aPPGT substrate alone in vivo, both under normal conditions and in an infarct model in mice, and found that there was a mild foreign body response with good integration of the substrate into the epicardial surface in mice hearts. We next studied the fabrication and properties of MCs by culturing mouse embryonic cardiomyocytes on the aPPGT substrate. To achieve myocardial-like concentrically contractile constructs, cocultures with supportive stromal cells were found to be essential and both mouse heart-derived stromal cells or bone-derived mouse mesenchymal stromal progenitor cells (mMSCs) could be used. These different stromal cell types produced MCs with different properties. The average beating rate of the constructs formed from mouse heart-derived stromal cells was significantly higher those constructs formed using mMSCs. Conversely, the constructs formed using mMSCs had reduced fibrotic extracellular matrix secretion and increased hepatocyte growth factor expression. Both of these mMSC construct properties may enhance integration and therapeutic efficacy of the construct postimplantation on the surface of the infarcted heart. This study thus demonstrates the formation of MCs using mechanically tailored aPPGT substrate and also demonstrates the effects of different stromal cell populations have on the properties of the resultant MCs, both of which are critical for future applications of tissue engineering in heart failure patients.


Asunto(s)
Módulo de Elasticidad , Infarto del Miocardio/terapia , Polímeros/química , Glicoles de Propileno/química , Ingeniería de Tejidos/métodos , Animales , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones , Miocardio/metabolismo
7.
Methods Mol Biol ; 698: 89-106, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21431513

RESUMEN

Clinical grade human mesenchymal stem cells (MSC) are manufactured and used in clinical trials for a range of regenerative and inflammatory diseases. Human MSC have now been derived from tissues other than bone marrow, such as placenta, as described in this laboratory protocol. It provides instructions for clinical grade MSC manufacturing according to the Code of Good Manufacturing Practice (cGMP) principles and according to policies and procedures of our internal Quality Management System (QMS), which is based on the International Organization for Standardization (ISO) standard requirements. Relevant organizational structure and QMS elements are presented.


Asunto(s)
Materiales Manufacturados/normas , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Placenta/citología , Animales , Técnicas de Cultivo de Célula , Núcleo Celular/metabolismo , Separación Celular , Criopreservación , Femenino , Citometría de Flujo , Humanos , Fenotipo , Embarazo , Etiquetado de Productos , Esterilización
8.
Stem Cells Dev ; 20(1): 77-87, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20446813

RESUMEN

Human mesenchymal stromal cells (hMSCs) have generated significant interest due to their potential use in clinical applications. hMSCs are present at low frequency in vivo, but after isolation can be expanded considerably, generating clinically useful numbers of cells. In this study, we demonstrate the use of a defined embryonic stem cell expansion medium, mTeSR (Stem Cell Technologies), for the expansion of bone-marrow-derived hMSCs. The hMSCs grow at comparable rates, demonstrate tri-lineage differentiation potential, and show similar surface marker profiles (CD29(+), CD44(+), CD49a(+), CD73(+), CD90(+), CD105(+), CD146(+), CD166(+), CD34(-), and CD45(-)) in both the fetal bovine serum (FBS)-supplemented medium and mTeSR. However, expression of early differentiation transcription factors runt-related transcription factor 2, sex-determining region Y box 9, and peroxisome proliferator-activated receptor gamma changed significantly. Both runt-related transcription factor 2 and sex-determining region Y box 9 were upregulated, whereas peroxisome proliferator-activated receptor gamma was downregulated in mTeSR compared with FBS. Although osteogenic and chondrogenic differentiation was comparable in cells grown in mTeSR compared to FBS, adipogenic differentiation was significantly decreased in mTeSR-expanded cells, both in terms of gene expression and absolute numbers of adipocytes. The removal of the FBS from the medium and the provision of a defined medium with disclosed composition make mTeSR a superior study platform for hMSC biology in a controlled environment. Further, this provides a key step toward generating a clinical-grade medium for expansion of hMSCs for clinical applications that rely on osteo- and chondroinduction of MSCs, such as bone repair and cartilage generation.


Asunto(s)
Condrogénesis/efectos de los fármacos , Medios de Cultivo/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Adolescente , Adulto , Animales , Antígenos de Superficie/metabolismo , Bovinos , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Condrogénesis/genética , Ensayo de Unidades Formadoras de Colonias , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Donantes de Tejidos , Factores de Transcripción/metabolismo , Adulto Joven
9.
Biomaterials ; 31(31): 7937-47, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20688386

RESUMEN

As strategies for manipulating cellular behaviour in vitro and in vivo become more sophisticated, synthetic biomaterial substrates capable of reproducing critical biochemical and biophysical properties (or cues) of tissue micro-environments will be required. Cytoskeletal tension has been shown to be highly deterministic of cell fate decisions, yet few synthetic biomaterials are capable of modulating cytoskeletal tension of adhered cells through variations in stiffness, at least in the ranges applicable to tissue properties (e.g., 1-100 kPa), whilst also possessing other required properties, such as biodegradability, biocompatibility and processability. In this paper we describe a non-cytotoxic polymer system based on acrylated polypropylene glycol triol (aPPGT). This new elastomer system has tunable elastic moduli, is degradable, can be easily surface modified and can be manufactured into porous three dimensional scaffolds or micropatterned substrates. We demonstrate that the PPGT substrates can modulate hMSC morphology, growth, and differentiation, and that they can produce similar outcomes as observed for a non-degradable polyacrylamide substrate, confirming their utility as a degradable elastomer for tissue engineering and other biomedical applications.


Asunto(s)
Acrilatos/farmacología , Módulo de Elasticidad/efectos de los fármacos , Elastómeros/farmacología , Polímeros/farmacología , Glicoles de Propileno/farmacología , Ingeniería de Tejidos/métodos , Acrilatos/química , Adipogénesis/efectos de los fármacos , Adolescente , Adulto , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/farmacología , Reactivos de Enlaces Cruzados/farmacología , Elastómeros/química , Humanos , Microscopía Electrónica de Rastreo , Persona de Mediana Edad , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/metabolismo , Osteogénesis/efectos de los fármacos , Polímeros/química , Glicoles de Propileno/química , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Propiedades de Superficie/efectos de los fármacos , Andamios del Tejido/química , Factores de Transcripción/metabolismo , Adulto Joven
10.
Cell Transplant ; 19(1): 29-42, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19878627

RESUMEN

Chondrogenesis of mesenchymal stem cells (MSCs) is typically induced when they are condensed into a single aggregate and exposed to transforming growth factor-beta (TGF-beta). Hypoxia, like aggregation and TGF-beta delivery, may be crucial for complete chondrogenesis. However, the pellet dimensions and associated self-induced oxygen gradients of current chondrogenic methods may limit the effectiveness of in vitro differentiation and subsequent therapeutic uses. Here we describe the use of embryoid body-forming technology to produce microscopic aggregates of human bone marrow MSCs (BM-MSCs) for chondrogenesis. The use of micropellets reduces the formation of gradients within the aggregates, resulting in a more homogeneous and controlled microenvironment. These micropellet cultures (approximately 170 cells/micropellet) as well as conventional pellet cultures (approximately 2 x 10(5) cells/pellet) were chondrogenically induced under 20% and 2% oxygen environments for 14 days. Compared to conventional pellets under both environments, micropellets differentiated under 2% O(2) showed significantly increased sulfated glycosaminoglycan (sGAG) production and more homogeneous distribution of proteoglycans and collagen II. Aggrecan and collagen II gene expressions were increased in pellet cultures differentiated under 2% O(2) relative to 20% O(2) pellets but 2% O(2) micropellets showed even greater increases in these genes, as well as increased SOX9. These results suggest a more advanced stage of chondrogenesis in the micropellets accompanied by more homogeneous differentiation. Thus, we present a new method for enhancing MSC chondrogenesis that reveals a unique relationship between oxygen tension and aggregate size. The inherent advantages of chondrogenic micropellets over a single macroscopic aggregate should allow for easy integration with a variety of cartilage engineering strategies.


Asunto(s)
Diferenciación Celular/fisiología , Condrocitos/metabolismo , Condrogénesis/fisiología , Hipoxia/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Agrecanos/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Agregación Celular/fisiología , Técnicas de Cultivo de Célula/métodos , Linaje de la Célula/fisiología , Proliferación Celular , Separación Celular/métodos , Células Cultivadas , Condrocitos/citología , Colágeno Tipo II/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Hipoxia/fisiopatología , Células Madre Mesenquimatosas/citología , Consumo de Oxígeno/fisiología , Esferoides Celulares/citología , Esferoides Celulares/metabolismo
11.
Tissue Eng Part C Methods ; 16(3): 407-15, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19622005

RESUMEN

In membrane bioreactors the cells are isolated from the bulk medium through a semipermeable membrane. This concept, which is analogous to how the circulatory system supplies solid tissues with nutrients, allows the maintenance of cells at much higher densities than is possible in traditional cultures. The membrane-based microbioreactor described herein is easy to operate, requiring only a pipette to load and harvest cells. A 10 microL culture volume was isolated from 1 mL of bulk medium through a semipermeable membrane having a molecular weight cutoff of 10 kDa. Here we describe the benefits regarding the retention of both cells and their secretions within this small culture volume using the following two model systems: hematopoietic stem cell expansion and mesenchymal stem cell-derived cartilage matrix accumulation.


Asunto(s)
Reactores Biológicos , Membranas Artificiales , Diferenciación Celular , Medios de Cultivo , Células Madre Hematopoyéticas/citología , Humanos
12.
Br J Haematol ; 144(4): 571-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19077161

RESUMEN

Mesenchymal stem cells (MSC) are being used increasingly in clinical trials for a range of regenerative and inflammatory diseases. Bone marrow is the traditional source but is relatively inaccessible in large volume. MSC have now been derived from tissues other than bone marrow including placenta and adipose tissue. We have used placenta obtained after delivery as a source of MSC and have been unable to detect any marked differences from marrow-derived MSC in terms of cell surface phenotype, chemokine receptor display, mesodermal differentiation capacity or immunosuppressive ability. This report described our manufacturing process for isolating and expanding placenta-derived human MSC and their safe infusion into the first patient in a clinical trial program of human placenta-derived MSC.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Placenta/citología , Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Separación Celular/métodos , Criopreservación/métodos , Resultado Fatal , Femenino , Citometría de Flujo/métodos , Humanos , Inmunofenotipificación , Leucemia Mieloide Aguda/terapia , Masculino , Células Madre Mesenquimatosas/inmunología , Resultado del Tratamiento , Adulto Joven
13.
Int J Cell Biol ; 2009: 906507, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20130765

RESUMEN

Cardiovascular diseases, particularly acute myocardial infarction, are the leading causes of death worldwide. Important advances have been made in the secondary treatment for cardiovascular diseases such as heart transplantation and medical and surgical therapies. Although these therapies alleviate symptoms, and may even improve survival, none can reverse the disease process and directly repair the lasting damage. Thus, the cure of cardiovascular diseases remains a major unmet medical need. Recently, cellular therapy has been proposed as a candidate treatment for this. Many stem and progenitor cell populations have each been suggested as a potential basis for such therapy. This review assesses some of the more notable exogenous adult cell candidates and provides insights into the mechanisms by which they may mediate improvement in cardiac function following acute myocardial infarction. Research into the cellular therapy field is of great importance for the further planning of clinical trials for cardiac cellular myoplasty.

14.
Int J Cell Biol ; 2009: 904682, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20130773

RESUMEN

Mesenchymal stem cells or multipotent mesenchymal stromal cells (both referred to as MSC) have been shown in some studies to have a beneficial effect on myocardial recovery after infarct. Current strategies for MSC delivery to heart involve intravenous, intraarterial, and intramuscular delivery. Different routes of MSC delivery and a lack of knowledge of the mechanisms that MSC utilise to migrate in vivo has most likely led to the marked variations in results that have been found. This review aims to summarise the current knowledge of MSC migratory mechanisms and looks to future methods of MSC manipulation prior to delivery in order to enhance MSC migration and engraftment.

15.
Stem Cells Dev ; 17(6): 1095-107, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19006451

RESUMEN

Bone marrow is the traditional source of human multipotent mesenchymal stem cells (MSCs), but placenta appears to be an alternative and more readily available source. This study comprehensively compared human placenta-derived MSC (hpMSC) and human bone marrow-derived MSC (hbmMSC) in terms of cell characteristics, optimal growth conditions and in vivo safety specifically to determine if hpMSC could represent a source of human MSC for clinical trial. MSC were isolated from human placenta (hpMSC) and human bone marrow (hbmMSC) and expanded ex vivo using good manufacturing practice-compliant reagents. hpMSC and hbmMSC showed similar proliferation characteristics in different basal culture media types, fetal calf serum (FCS) concentrations, FCS heat-inactivation experiments, flask types and media replacement responsiveness. However, hpMSC and hbmMSC differed with respect to their proliferation capabilities at different seeding densities, with hbmMSC proliferating more slowly than hpMSC in every experiment. hpMSC had greater long-term growth ability than hbmMSC. MSC from both sources exhibited similar light microscopy morphology, size, cell surface phenotype, and mesodermal differentiation ability with the exception that hpMSC consistently appeared less able to differentiate to the adipogenic lineage. A comparison of both hbmMSC and hpMSC from early and medium passage cultures using single-nucleotide polymorphism (SNP) GeneChip analysis confirmed GTG-banding data that no copy number changes had been acquired during sequential passaging. In three of three informative cases (in which the gender of the delivered baby was male), hpMSC were of maternal origin. Neither hpMSC nor hbmMSC caused any acute toxicity in normal mice when injected intravenously at the same, or higher, doses than those currently used in clinical trials of hbmMSC. This study suggests that human placenta is an acceptable alternative source for human MSC and their use is currently being evaluated in clinical trials.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular/fisiología , Proliferación Celular , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Placenta/citología , Adulto , Animales , Células de la Médula Ósea/metabolismo , Separación Celular/métodos , Células Cultivadas , Femenino , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Células Madre Multipotentes/metabolismo , Placenta/metabolismo , Factores de Tiempo
16.
Stem Cells Dev ; 17(5): 929-40, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18564033

RESUMEN

We compared potential trafficking mechanisms used by human (h) multipotent mesenchymal stem cells (MSC) derived from bone marrow (bm) or placenta (p). Both hbmMSC and hpMSC expressed a broad range of cell surface adhesion molecules including beta1-integrins (CD29) and CD44. Array data showed that both hbmMSC and hpMSC expressed mRNA for the cell adhesion molecules CD54 (ICAM-1), E-cadherin, CD166 (ALCAM), CD56 (NCAM), CD106 (VCAM-1), CD49a, b, c, e and f (integrins alpha1, 2, 3, 4 and 6), integrin alpha11, CD51 (integrin alphaV), and CD29 (integrins beta1). Functional binding of hpMSC, but not hbmMSC to VCAM-1 was demonstrated using recombinant chimeric constructs. Neither bone marrow nor placental MSC expressed ligands to endothelial selectins such as PSGL-1 or sialyl Lewis X (sLe(x)) carbohydrates and neither were able to bind functionally to chimeric constructs of the endothelial selectins CD62E (E-selectin) and CD62P (P-selectin). Furthermore, MSC expressed a restricted range of transferases necessary for expression of sLe(x), with no detectable expression of fucosyl transferases IV or VII. Placental MSC, but not hbmMSC, expressed mRNA for the chemokine receptors CCR1 and CCR3, and both hbmMSC and hpMSC expressed mRNA for CCR7, CCR8, CCR10, CCR11, CXCR4 and CXCR6. Intracellular chemokine receptor protein expression of CCR1, CCR3, CXCR3, CXCR4 and CXCR6 was detected in both hbmMSC and hpMSC. Cell surface expression of chemokine receptors was much more restricted with only CXCR6 displaying a strong signal on hbmMSC and hpMSC. Although cell surface expression of CXCR4 was not detected, MSC migrated in response to its ligand, CXCL12 (SDF-1). Thus, hbmMSC and hpMSC have an almost identical profile for cell surface adhesion and chemokine receptor molecules at the mRNA and protein levels. However, at the functional level, hpMSC likely utilise VLA-4-mediated binding in a superior manner to hbmMSC and thus may have superior engraftment properties to hbmMSC in vivo.


Asunto(s)
Células de la Médula Ósea/citología , Movimiento Celular , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Placenta/citología , Adolescente , Adulto , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/farmacología , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Femenino , Citometría de Flujo , Fucosa/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glicosiltransferasas/metabolismo , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/enzimología , Mesodermo/citología , Mesodermo/efectos de los fármacos , Células Madre Multipotentes/efectos de los fármacos , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Selectinas/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
17.
Transfus Med Hemother ; 35(4): 279-285, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-21512643

RESUMEN

SUMMARY: Therapeutic applications of cells are likely to increase greatly in the future. Cell and cell-based gene therapy manufacturing facilities need to be purpose-designed and accredited by their national medicinal regulatory body. Production scientists need to work in close tandem with quality assurance and ethics committees to absolutely ensure the safety of new cellular products. In this review, we consider the need for preclinical safety and efficacy data, tissue source for manufacture of clinical grade human mesenchymal stem cells, aseptic tissue processing, indemnification, and the role of the national medicinal regulatory body in appropriate clinical trial design.

18.
Semin Cell Dev Biol ; 18(6): 846-58, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18024097

RESUMEN

Mesenchymal stromal cells (MSC) are multipotent cells that can be derived from many different organs and tissues. They have been demonstrated to play a role in tissue repair and regeneration in both preclinical and clinical studies. They also have remarkable immunosuppressive properties. We describe their application in settings that include the cardiovascular, central nervous, gastrointestinal, renal, orthopaedic and haematopoietic systems. Manufacturing of MSC for clinical trials is also discussed. Since tissue matching between MSC donor and recipient does not appear to be required, MSC may be the first cell type able to be used as an "off-the-shelf" therapeutic product.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Células del Estroma/citología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Células Madre Multipotentes/citología , Células Madre Multipotentes/fisiología , Regeneración , Células del Estroma/fisiología , Células del Estroma/trasplante
19.
Tissue Eng ; 13(4): 819-29, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17371154

RESUMEN

The demand for treatment strategies for damaged musculoskeletal tissue is continuously growing, especially with the increasing number of older people with degenerative diseases of the skeletal system such as osteoarthritis (OA). Because depletion of multipotent cells has been implicated in degenerative joint diseases, cell-based therapies have been proposed for tissue regeneration, especially for cartilage repair. The aim of the present study is to focus on the possibility of deriving and expanding multipotential mesenchymal stem cells (MSCs) from bone marrow samples of patients with OA by characterizing MSCs at the single cell level. Single-cell clonal cultures were established in 96-well plates by limiting dilution of bone marrow stromal cells (BMSCs) from three patients with OA. Fourteen clones were established for subsequent characterization. There was a wide variation in cell doubling times, with the time taken to reach 20 population doublings ranging from 37 days to more than 100 days. The clones were grouped into fast-growing and slow-growing clones. All except one of the fast-growing stem cell clones were tripotential. However, the slow-growing clones showed limited differentiation potential and morphological changes associated with cellular senescence with extended duration in culture. Flow cytometric analysis indicated a strong need to investigate for novel cell-surface characteristic markers of BMSCs because there was no obvious difference in the expression of the selected characteristic BMSC cell surface markers CD29, CD44, CD90, CD105, and CD166 between fast-growing and slow-growing clones. This study has demonstrated the existence of a fast-growing multipotential MSC population from bone marrow samples of patients with OA. Therefore, despite a supposedly smaller stem cell compartment in these patients, we demonstrate here that they can still yield a potentially therapeutically useful source of syngeneic MSCs.


Asunto(s)
Adipocitos/patología , Células Madre Adultas/patología , Células de la Médula Ósea/patología , Condrocitos/patología , Osteoartritis de la Rodilla/patología , Osteoblastos/patología , Diferenciación Celular , Separación Celular/métodos , Células Cultivadas , Clonación de Organismos/métodos , Humanos , Células del Estroma/patología
20.
J Acoust Soc Am ; 122(5): 2560-70, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18189547

RESUMEN

This article examines the effects of spatial field shifts in ocean acoustic environmental sensitivity analysis. Acoustic sensitivity studies are typically based on comparing acoustic fields computed for a reference environmental model and for a perturbed model in which one or more parameters have been changed. The perturbation to the acoustic field due to the perturbed environment generally includes a component representing a spatial shift of the field (i.e., local field structure remains coherent, but shifts in range and/or depth) and a component representing a change to the shifted field. Standard sensitivity measures based on acoustic perturbations at a fixed point can indicate high sensitivity in cases where the field structure changes very little, but is simply shifted by a small spatial offset; this can conflict with an intuitive understanding of sensitivity. This article defines and compares fixed-point and field-shift corrected sensitivity measures. The approaches are illustrated with examples of deterministic sensitivity (i.e., sensitivity to a specific environmental change) and stochastic sensitivity (sensitivity to environmental uncertainty) in range-independent and range-dependent environments.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...