Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Biomed Mater Res A ; 100(12): 3276-86, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22733596

ABSTRACT

Scaffolds with aligned channels based on acrylate copolymers, which had previously demonstrated good compatibility with neural progenitor cells were studied as colonizable structures both in vitro with neural progenitor cells and in vivo, implanted without cells in two different locations, in the cortical plate of adult rat brains and close to the subventricular zone. In vitro, neuroprogenitors colonize the scaffold and differentiate into neurons and glia within its channels. When implanted in vivo immunohistochemical analysis by confocal microscopy for neural and endothelial cells markers demonstrated that the scaffolds maintained continuity with the surrounding neural tissue and were colonized by GFAP-positive cells and, in the case of scaffolds implanted in contact with the subventricular zone, by neurons. Local angiogenesis was evidenced in the interior of the scaffolds' pores. New axons and neural cells from the adult neural niche abundantly colonized the biomaterial's inner structure after 2 months, and minimal scar formation was manifest around the implant. These findings indicate the biocompatibility of the polymeric material with the brain tissue and open possibilities to further studies on the relevance of factors such as scaffold structure, scaffold seeding and scaffold placement for their possible use in regenerative strategies in the central nervous system. The development of neural interfaces with minimized glial scar and improved tissue compatibility of the implants may also benefit from these results. © 2012 Wiley Periodicals, Inc. J Biomed Mater Res Part A 100A:3276-3286, 2012.


Subject(s)
Aging/physiology , Brain/physiology , Implants, Experimental , Tissue Scaffolds/chemistry , Adult Stem Cells/cytology , Animals , Cell Differentiation , Cerebral Cortex/physiology , Female , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Neovascularization, Physiologic , Neural Stem Cells/cytology , Neuroglia/cytology , Neurons/cytology , Rats , Rats, Wistar , Stem Cell Transplantation
2.
Cell Transplant ; 21(5): 1023-37, 2012.
Article in English | MEDLINE | ID: mdl-22305117

ABSTRACT

Fresh adipose-derived cells have been shown to be effective in the treatment of acute myocardial infarction (MI), but their role in the chronic setting is unknown. We sought to determine the long-term effect of the adipose derived-stromal vascular fraction (SVF) cell transplantation in a rat model of chronic MI. MI was induced in 82 rats by permanent coronary artery ligation and 5 weeks later rats were allocated to receive an intramyocardial injection of 10(7) GFP-expressing fresh SVF cells or culture media as control. Heart function and tissue metabolism were determined by echocardiography and (18)F-FDG-microPET, respectively, and histological studies were performed for up to 3 months after transplantation. SVF induced a statistically significant long-lasting (3 months) improvement in cardiac function and tissue metabolism that was associated with increased revascularization and positive heart remodeling, with a significantly smaller infarct size, thicker infarct wall, lower scar fibrosis, and lower cardiac hypertrophy. Importantly, injected cells engrafted and were detected in the treated hearts for at least 3 months, directly contributing to the vasculature and myofibroblasts and at negligible levels to cardiomyocytes. Furthermore, SVF release of angiogenic (VEGF and HGF) and proinflammatory (MCP-1) cytokines, as well as TIMP1 and TIMP4, was demonstrated in vitro and in vivo, strongly suggesting that they have a trophic effect. These results show the potential of SVF to contribute to the regeneration of ischemic tissue and to provide a long-term functional benefit in a rat model of chronic MI, by both direct and indirect mechanisms.


Subject(s)
Adipocytes/cytology , Myocardial Infarction/therapy , Paracrine Communication , Stromal Cells/transplantation , Ventricular Remodeling , Angiogenic Proteins/metabolism , Animals , Cell Differentiation , Chronic Disease , Cytokines/metabolism , Disease Models, Animal , Echocardiography , Female , Heart Ventricles/physiopathology , Myocardial Infarction/pathology , Myocardial Revascularization , Phenotype , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Stromal Cells/cytology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinase-4
3.
Micron ; 43(5): 589-99, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22227011

ABSTRACT

GFP has emerged as an important reporter for monitoring gene expression, protein localization, cell transformation and cell lineage. The development of GFP as a marker in many different biological systems has emphasized the need to image GFP at high resolution. GFP immunogold labeling with colloidal gold particles becomes essential for electron microscopy (EM) ultrastructural detection. Because of the small size, colloidal gold particles require silver enhancement, a procedure to increase the size of the particle as well as gold toning to stabilize the silver layer. GFP preembedding immunogold staining enables high quality cellular-ultrastructural EM analysis mainly for two reasons, on one hand it allows adequate fixation for EM analysis maintaining GFP antigenicity, on the other hand it also enables the epoxy resins inclusion after immunogold staining. Both of them help to preserve better the ultrastructure. However GFP immunogold staining presents some drawbacks, such as the progressive decrease in immunogold labeling with tissue depth. Special attention must be taken when using GFP-tagged protein, since the fusion could interfere with their localization and function. In this review we provide a detailed protocol of the GFP immunogold staining, their main applications for EM and possible troubles.


Subject(s)
Fluorescent Dyes , Green Fluorescent Proteins , Immunohistochemistry/methods , Staining and Labeling/methods , Animals , Humans
4.
Cell Transplant ; 19(8): 919-36, 2010.
Article in English | MEDLINE | ID: mdl-20350351

ABSTRACT

The ultimate therapy for ischemic stroke is restoration of blood supply in the ischemic region and regeneration of lost neural cells. This might be achieved by transplanting cells that differentiate into vascular or neuronal cell types, or secrete trophic factors that enhance self-renewal, recruitment, long-term survival, and functional integration of endogenous stem/progenitor cells. Experimental stroke models have been developed to determine potential beneficial effect of stem/progenitor cell-based therapies. To follow the fate of grafted cells in vivo, a number of noninvasive imaging approaches have been developed. Magnetic resonance imaging (MRI) is a high-resolution, clinically relevant method allowing in vivo monitoring of cells labeled with contrast agents. In this study, labeling efficiency of three different stem cell populations [mouse embryonic stem cells (mESC), rat multipotent adult progenitor cells (rMAPC), and mouse mesenchymal stem cells (mMSC)] with three different (ultra)small superparamagnetic iron oxide [(U)SPIO] particles (Resovist, Endorem, Sinerem) was compared. Labeling efficiency with Resovist and Endorem differed significantly between the different stem cells. Labeling with (U)SPIOs in the range that allows detection of cells by in vivo MRI did not affect differentiation of stem cells when labeled with concentrations of particles needed for MRI-based visualization. Finally, we demonstrated that labeled rMAPC could be detected in vivo and that labeling did not interfere with their migration. We conclude that successful use of (U)SPIOs for MRI-based visualization will require assessment of the optimal (U)SPIO for each individual (stem) cell population to ensure the most sensitive detection without associated toxicity.


Subject(s)
Contrast Media , Embryonic Stem Cells/cytology , Magnetic Resonance Imaging , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Animals , Cell Differentiation , Cell Movement , Dextrans , Female , Flow Cytometry , Magnetite Nanoparticles , Male , Mice , Multipotent Stem Cells/transplantation , Phenotype , Rats , Stroke/therapy
5.
J Neurosci ; 24(49): 11171-81, 2004 Dec 08.
Article in English | MEDLINE | ID: mdl-15590934

ABSTRACT

The subventricular zone (SVZ) is one of the sources of adult neural stem cells (ANSCs) in the mouse brain. Precursor cells proliferate in the SVZ and migrate through the rostral migratory stream (RMS) to the olfactory bulb (OB), where they differentiate into granule and periglomerular cells. Few transcription factors are known to be responsible for regulating NSC proliferation, migration, and differentiation processes; even fewer have been found to be responsible for the organization of the SVZ and RMS. For this reason, we studied the ventral anterior homeobox (Vax1) gene in NSC proliferation and in SVZ organization. We found that Vax1 is strongly expressed in the SVZ and in the RMS and that, in the absence of Vax1, embryonic precursor cells proliferate 100 times more than wild-type controls, in vitro. The SVZ of Vax1(-/-) brains is hyperplastic and mostly disorganized, and the RMS is missing, causing a failure of precursor cell migration to the OBs, which as a result are severely hypoplastic. Moreover, we found that Vax1 is essential for the correct differentiation of ependyma and astrocytes. Together, these data indicate that Vax1 is a potent regulator of SVZ organization and NSC proliferation, with important consequences on postnatal neurogenesis.


Subject(s)
Genes, Homeobox/physiology , Homeodomain Proteins/physiology , Neuropeptides/physiology , Stem Cells/physiology , Telencephalon/growth & development , Telencephalon/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuropeptides/biosynthesis , Neuropeptides/genetics , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , Telencephalon/cytology , Telencephalon/embryology , Transcription Factors/metabolism
6.
J Neurosci ; 22(6): 2255-64, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11896165

ABSTRACT

The subventricular zone (SVZ) is the largest germinal layer in the adult mammalian brain and comprises stem cells, transit-amplifying progenitors, and committed neuroblasts. Although the SVZ contains the highest concentration of dividing cells in the adult brain, the intracellular mechanisms controlling their proliferation have not been elucidated. We show here that loss of the cyclin-dependent kinase inhibitor p27Kip1 has very specific effects on a population of CNS progenitors responsible for adult neurogenesis. Using bromodeoxyuridine and [(3)H]thymidine incorporation to label cells in S phase and cell-specific markers and electron microscopy to identify distinct cell types, we compared the SVZ structure and proliferation characteristics of wild-type and p27Kip1-null mice. Loss of p27Kip1 had no effect on the number of stem cells but selectively increased the number of the transit-amplifying progenitors concomitantly with a reduction in the number of neuroblasts. We conclude that cell-cycle regulation of SVZ adult progenitors is remarkably cell-type specific, with p27Kip1 being a key regulator of the cell division of the transit-amplifying progenitors.


Subject(s)
Neurons/cytology , Stem Cells/cytology , Stem Cells/physiology , Tumor Suppressor Proteins/deficiency , Animals , Apoptosis/genetics , Bromodeoxyuridine/metabolism , Cell Count , Cell Cycle , Cell Cycle Proteins/genetics , Cell Differentiation/drug effects , Cell Division/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27 , Immunohistochemistry , In Situ Nick-End Labeling , Lateral Ventricles/cytology , Lateral Ventricles/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Spheroids, Cellular/cytology , Stem Cells/metabolism , Thymidine/metabolism , Tumor Suppressor Proteins/genetics
7.
J Neurosci ; 22(2): 437-45, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11784788

ABSTRACT

The lateral walls of the forebrain lateral ventricles are the richest source of stem cells in the adult mammalian brain. These stem cells give rise to new olfactory neurons that are renewed throughout life. The neurons originate in the subventricular zone (SVZ), migrate within the rostral extension (RE) of the SVZ along the rostral migratory stream (RMS) within tube-like structures formed of glial cells, to eventually reach the olfactory bulb (OB). We demonstrate that, contrary to the current view, multipotential (neuronal-astroglial-oligodendroglial) precursors with stem cell features can be isolated not only from the SVZ but also from the entire RE, including the distal portion within the OB. Specifically, these stem cells do not derive from the migratory neuroblasts coming from the SVZ. Interestingly, stem cells isolated from the proximal RE generate significantly more oligodendrocytes, and those from the distal RE proliferate significantly more slowly than stem cells derived from the SVZ and other RE regions. These findings demonstrate that stem cells are not confined to the forebrain periventricular region and indicate that stem cells endowed with different functional characteristics occur at different levels of the SVZ-RE pathway.


Subject(s)
Neurons/cytology , Olfactory Bulb/cytology , Stem Cells/cytology , Animals , Astrocytes/cytology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Division/drug effects , Cell Line/cytology , Cell Movement/physiology , Cell Separation , Cells, Cultured , Clone Cells/classification , Clone Cells/cytology , Clone Cells/drug effects , Growth Substances/pharmacology , Lateral Ventricles/cytology , Mice , Neurons/classification , Neurons/metabolism , Neurotransmitter Agents/metabolism , Oligodendroglia/cytology , Phenotype , Spheroids, Cellular/cytology , Stem Cells/classification , Stem Cells/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...