Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Cell Mol Med ; 16(1): 107-17, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21418520

RESUMEN

We recently developed a method to control the in vivo distribution of vascular endothelial growth factor (VEGF) by high throughput Fluorescence-Activated Cell Sorting (FACS) purification of transduced progenitors such that they homogeneously express specific VEGF levels. Here we investigated the long-term safety of this method in chronic hind limb ischemia in nude rats. Primary myoblasts were transduced to co-express rat VEGF-A(164) (rVEGF) and truncated ratCD8a, the latter serving as a FACS-quantifiable surface marker. Based on the CD8 fluorescence of a reference clonal population, which expressed the desired VEGF level, cells producing similar VEGF levels were sorted from the primary population, which contained cells with very heterogeneous VEGF levels. One week after ischemia induction, 12 × 10(6) cells were implanted in the thigh muscles. Unsorted myoblasts caused angioma-like structures, whereas purified cells only induced normal capillaries that were stable after 3 months. Vessel density was doubled in engrafted areas, but only approximately 0.1% of muscle volume showed cell engraftment, explaining why no increase in total blood flow was observed. In conclusion, the use of FACS-purified myoblasts granted the cell-by-cell control of VEGF expression levels, which ensured long-term safety in a model of chronic ischemia. Based on these results, the total number of implanted cells required to achieve efficacy will need to be determined before a clinical application.


Asunto(s)
Separación Celular/métodos , Miembro Posterior/irrigación sanguínea , Isquemia/fisiopatología , Mioblastos/fisiología , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Biomarcadores/metabolismo , Antígenos CD8/genética , Antígenos CD8/metabolismo , Trasplante de Células , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/citología , Mioblastos/trasplante , Ratas , Ratas Desnudas , Factor A de Crecimiento Endotelial Vascular/genética
2.
Stem Cells Transl Med ; 5(12): 1730-1738, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27460852

RESUMEN

: Chondrogenic differentiation of bone marrow-derived mesenchymal stromal/stem cells (MSCs) can be induced by presenting morphogenetic factors or soluble signals but typically suffers from limited efficiency, reproducibility across primary batches, and maintenance of phenotypic stability. Considering the avascular and hypoxic milieu of articular cartilage, we hypothesized that sole inhibition of angiogenesis can provide physiological cues to direct in vivo differentiation of uncommitted MSCs to stable cartilage formation. Human MSCs were retrovirally transduced to express a decoy soluble vascular endothelial growth factor (VEGF) receptor-2 (sFlk1), which efficiently sequesters endogenous VEGF in vivo, seeded on collagen sponges and immediately implanted ectopically in nude mice. Although naïve cells formed vascularized fibrous tissue, sFlk1-MSCs abolished vascular ingrowth into engineered constructs, which efficiently and reproducibly developed into hyaline cartilage. The generated cartilage was phenotypically stable and showed no sign of hypertrophic evolution up to 12 weeks. In vitro analyses indicated that spontaneous chondrogenic differentiation by blockade of angiogenesis was related to the generation of a hypoxic environment, in turn activating the transforming growth factor-ß pathway. These findings suggest that VEGF blockade is a robust strategy to enhance cartilage repair by endogenous or grafted mesenchymal progenitors. This article outlines the general paradigm of controlling the fate of implanted stem/progenitor cells by engineering their ability to establish specific microenvironmental conditions rather than directly providing individual morphogenic cues. SIGNIFICANCE: Chondrogenic differentiation of mesenchymal stromal/stem cells (MSCs) is typically targeted by morphogen delivery, which is often associated with limited efficiency, stability, and robustness. This article proposes a strategy to engineer MSCs with the capacity to establish specific microenvironmental conditions, supporting their own targeted differentiation program. Sole blockade of angiogenesis mediated by transduction for sFlk-1, without delivery of additional morphogens, is sufficient for inducing MSC chondrogenic differentiation. The findings represent a relevant step forward in the field because the method allowed reducing interdonor variability in MSC differentiation efficiency and, importantly, onset of a stable, nonhypertrophic chondrocyte phenotype.


Asunto(s)
Células de la Médula Ósea/citología , Condrogénesis , Células Madre Mesenquimatosas/citología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Hipertrofia , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Oxígeno/farmacología , Transducción de Señal/efectos de los fármacos , Transducción Genética , Factor de Crecimiento Transformador beta/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
3.
Biomaterials ; 34(21): 5025-35, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23566801

RESUMEN

Rapid vascularisation of tissue-engineered osteogenic grafts is a major obstacle in the development of regenerative medicine approaches for bone repair. Vascular endothelial growth factor (VEGF) is the master regulator of vascular growth. We investigated a cell-based gene therapy approach to generate osteogenic grafts with an increased vascularization potential in an ectopic nude rat model in vivo, by genetically modifying human bone marrow-derived stromal/stem cells (BMSC) to express rat VEGF. BMSC were loaded onto silicate-substituted apatite granules, which are a clinically established osteo-conductive material. Eight weeks after implantation, the vascular density of constructs seeded with VEGF-BMSC was 3-fold greater than with control cells, consisting of physiologically structured vascular networks with both conductance vessels and capillaries. However, VEGF specifically caused a global reduction in bone quantity, which consisted of thin trabeculae of immature matrix. VEGF did not impair BMSC engraftment in vivo, but strongly increased the recruitment of TRAP- and Cathepsin K-positive osteoclasts. These data suggest that VEGF over-expression is effective to improve the vascularization of osteogenic grafts, but also has the potential to disrupt bone homoeostasis towards excessive degradation, posing a challenge to its clinical application in bone tissue engineering.


Asunto(s)
Resorción Ósea/patología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica , Osteogénesis , Factor A de Crecimiento Endotelial Vascular/metabolismo , Fosfatasa Ácida/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Matriz Ósea/metabolismo , Catepsina K/metabolismo , Proliferación Celular , Supervivencia Celular , Humanos , Isoenzimas/metabolismo , Células Madre Mesenquimatosas/citología , Osteoclastos/patología , Ratas , Fosfatasa Ácida Tartratorresistente
4.
Tissue Eng Part C Methods ; 18(4): 283-92, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22070632

RESUMEN

Adult mesenchymal stromal/stem cells (MSCs) are a valuable source of multipotent progenitors for tissue engineering and regenerative medicine, but may require to be genetically modified to widen their efficacy in therapeutic applications. For example, overexpression of the angiogenic factor vascular endothelial growth factor (VEGF) at controlled levels is an attractive strategy to overcome the crucial bottleneck of graft vascularization and to avoid aberrant vascular growth. Since the regenerative potential of MSCs is rapidly lost during in vitro expansion, we sought to develop an optimized technique to achieve high-efficiency retroviral vector transduction of MSCs derived from both adipose tissue (adipose stromal cells, ASCs) or bone marrow (BMSCs) and rapidly select cells expressing desired levels of VEGF with minimal in vitro expansion. The proliferative peak of freshly isolated human ASCs and BMSCs was reached 4 and 6 days after plating, respectively. By performing retroviral vector transduction at this time point, >90% efficiency was routinely achieved before the first passage. MSCs were transduced with vectors expressing rat VEGF(164) quantitatively linked to a syngenic cell surface marker (truncated rat CD8). Retroviral transduction and VEGF expression did not affect MSC phenotype nor impair their in vitro proliferation and differentiation potential. Transgene expression was also maintained during in vitro differentiation. Furthermore, three subpopulations of transduced BMSCs homogeneously producing specific low, medium, and high VEGF doses could be prospectively isolated by flow cytometry based on the intensity of their CD8 expression already at the first passage. In conclusion, this optimized platform allowed the generation of populations of genetically modified MSCs, expressing specific levels of a therapeutic transgene, already at the first passage, thereby minimizing in vitro expansion and loss of regenerative potential.


Asunto(s)
Células Madre Adultas/citología , Separación Celular/métodos , Citometría de Flujo/métodos , Células Madre Mesenquimatosas/citología , Transducción Genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Vectores Genéticos/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratas , Retroviridae/genética , Transgenes/genética
5.
Hum Gene Ther Methods ; 23(5): 346-56, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23075102

RESUMEN

Vascular endothelial growth factor (VEGF) can induce normal angiogenesis or the growth of angioma-like vascular tumors depending on the amount secreted by each producing cell because it remains localized in the microenvironment. In order to control the distribution of VEGF expression levels in vivo, we recently developed a high-throughput fluorescence-activated cell sorting (FACS)-based technique to rapidly purify transduced progenitors that homogeneously express a specific VEGF dose from a heterogeneous primary population. Here we tested the hypothesis that cell-based delivery of a controlled VEGF level could induce normal angiogenesis in the heart, while preventing the development of angiomas. Freshly isolated human adipose tissue-derived stem cells (ASC) were transduced with retroviral vectors expressing either rat VEGF linked to a FACS-quantifiable cell-surface marker (a truncated form of CD8) or CD8 alone as control (CTR). VEGF-expressing cells were FACS-purified to generate populations producing either a specific VEGF level (SPEC) or uncontrolled heterogeneous levels (ALL). Fifteen nude rats underwent intramyocardial injection of 10(7) cells. Histology was performed after 4 weeks. Both the SPEC and ALL cells produced a similar total amount of VEGF, and both cell types induced a 50%-60% increase in both total and perfused vessel density compared to CTR cells, despite very limited stable engraftment. However, homogeneous VEGF expression by SPEC cells induced only normal and stable angiogenesis. Conversely, heterogeneous expression of a similar total amount by the ALL cells caused the growth of numerous angioma-like structures. These results suggest that controlled VEGF delivery by FACS-purified ASC may be a promising strategy to achieve safe therapeutic angiogenesis in the heart.


Asunto(s)
Expresión Génica , Miocardio/metabolismo , Neovascularización Fisiológica/genética , Factor A de Crecimiento Endotelial Vascular/genética , Adipocitos/citología , Adipocitos/metabolismo , Animales , Supervivencia Celular , Citometría de Flujo , Orden Génico , Vectores Genéticos/genética , Humanos , Inflamación , Masculino , Miocardio/patología , Neovascularización Patológica , Perfusión , Fenotipo , Ratas , Trasplante de Células Madre , Células Madre/citología , Células Madre/metabolismo , Transducción Genética , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda