Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 57(4): 1847-1862, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31845093

ABSTRACT

Understanding the mechanisms of glial scar formation by reactive astrocytes is crucial for elaborating a therapeutic strategy to brain and spinal cord injury. However, the extrinsic mechanisms that drive the polarization of reactive astrocytes, the first step in glial scar formation, remain poorly understood. Here, using an in vitro chemotaxis assay as an experimental model for polarization, we observed that Il4-M2 macrophages are stronger inducers of reactive astrocytes' polarization, compared to naive or M1 macrophages. Then, we showed that both ß1-integrin and Wnt/ß-catenin pathways in astrocytes are required for this polarization in vitro and in vivo after spinal cord crush injury in mice. These findings provide molecular targets for manipulating the polarization of reactive astrocytes in order to potentially enhance the healing of SCI lesions.


Subject(s)
Astrocytes/metabolism , Astrocytes/pathology , Cell Polarity , Macrophage Activation , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Wnt Signaling Pathway , Animals , Chemotaxis , Integrin beta1/metabolism , Mice, Inbred C57BL , Wnt3A Protein/metabolism
2.
Stem Cells Transl Med ; 8(3): 260-270, 2019 03.
Article in English | MEDLINE | ID: mdl-30485733

ABSTRACT

Tumorigenesis is an important problem that needs to be addressed in the field of human stem/progenitor cell transplantation for the treatment of subacute spinal cord injury (SCI). When certain "tumorigenic" cell lines are transplanted into the spinal cord of SCI mice model, there is initial improvement of motor function, followed by abrupt deterioration secondary to the effect of tumor growth. A significant proportion of the transplanted cells remains undifferentiated after transplantation and is thought to increase the risk of tumorigenesis. In this study, using lentiviral vectors, we introduced the herpes simplex virus type 1 thymidine kinase (HSVtk) gene into a human induced pluripotent stem cell-derived neural stem/progenitor cell (hiPSC-NS/PC) line that is known to undergo tumorigenic transformation. Such approach enables selective ablation of the immature proliferating cells and thereby prevents subsequent tumor formation. In vitro, the HSVtk system successfully ablated the immature proliferative neural cells while preserving mature postmitotic neuronal cells. Similar results were observed in vivo following transplantation into the injured spinal cords of immune-deficient (nonobese diabetic-severe combined immune-deficient) mice. Ablation of the proliferating cells exerted a protective effect on the motor function which was regained after transplantation, simultaneously defending the spinal cord from the harmful tumor growth. These results suggest a potentially promising role of suicide genes in opposing tumorigenesis during stem cell therapy. This system allows both preventing and treating tumorigenesis following hiPSC-NS/PC transplantation without sacrificing the improved motor function. Stem Cells Translational Medicine 2019;8:260&270.


Subject(s)
Carcinogenesis/pathology , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Spinal Cord Injuries/therapy , Spinal Cord/physiology , Animals , Cell Differentiation/physiology , Cells, Cultured , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neurons/physiology , Recovery of Function/physiology , Stem Cell Transplantation/methods
4.
Elife ; 62017 11 28.
Article in English | MEDLINE | ID: mdl-29179815

ABSTRACT

CHARGE syndrome is caused by heterozygous mutations in the chromatin remodeler, CHD7, and is characterized by a set of malformations that, on clinical grounds, were historically postulated to arise from defects in neural crest formation during embryogenesis. To better delineate neural crest defects in CHARGE syndrome, we generated induced pluripotent stem cells (iPSCs) from two patients with typical syndrome manifestations, and characterized neural crest cells differentiated in vitro from these iPSCs (iPSC-NCCs). We found that expression of genes associated with cell migration was altered in CHARGE iPSC-NCCs compared to control iPSC-NCCs. Consistently, CHARGE iPSC-NCCs showed defective delamination, migration and motility in vitro, and their transplantation in ovo revealed overall defective migratory activity in the chick embryo. These results support the historical inference that CHARGE syndrome patients exhibit defects in neural crest migration, and provide the first successful application of patient-derived iPSCs in modeling craniofacial disorders.


Subject(s)
CHARGE Syndrome/physiopathology , Cell Movement , Neural Crest/physiology , Animals , CHARGE Syndrome/genetics , Cell Differentiation , Cells, Cultured , Chick Embryo , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/physiology , Mutant Proteins/genetics , Mutation
6.
J Cell Biol ; 216(8): 2533-2550, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28642362

ABSTRACT

Understanding how the transcription factor signal transducer and activator of transcription-3 (STAT3) controls glial scar formation may have important clinical implications. We show that astrocytic STAT3 is associated with greater amounts of secreted MMP2, a crucial protease in scar formation. Moreover, we report that STAT3 inhibits the small GTPase RhoA and thereby controls actomyosin tonus, adhesion turnover, and migration of reactive astrocytes, as well as corralling of leukocytes in vitro. The inhibition of RhoA by STAT3 involves ezrin, the phosphorylation of which is reduced in STAT3-CKO astrocytes. Reduction of phosphatase and tensin homologue (PTEN) levels in STAT3-CKO rescues reactive astrocytes dynamics in vitro. By specific targeting of lesion-proximal, reactive astrocytes in Nestin-Cre mice, we show that reduction of PTEN rescues glial scar formation in Nestin-Stat3+/- mice. These findings reveal novel intracellular signaling mechanisms underlying the contribution of reactive astrocyte dynamics to glial scar formation.


Subject(s)
Astrocytes/enzymology , Cicatrix/enzymology , Neuroglia/enzymology , STAT3 Transcription Factor/metabolism , Spinal Cord Injuries/enzymology , Spinal Cord/enzymology , Wounds, Stab/enzymology , rho GTP-Binding Proteins/metabolism , Actomyosin/metabolism , Animals , Animals, Newborn , Astrocytes/pathology , Cell Adhesion , Cell Movement , Cells, Cultured , Cicatrix/genetics , Cicatrix/pathology , Coculture Techniques , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Genotype , Integrases/genetics , Macrophages/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Nestin/genetics , Neuroglia/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phenotype , Phosphorylation , Proteolysis , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Signal Transduction , Spinal Cord/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Transfection , Wounds, Stab/genetics , Wounds, Stab/pathology , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein
7.
PLoS One ; 10(8): e0136885, 2015.
Article in English | MEDLINE | ID: mdl-26317969

ABSTRACT

TCR-dependent and costimulation signaling, cell division, and cytokine environment are major factors driving cytokines expression induced by CD4(+) T cell activation. PEA-15 15 (Protein Enriched in Astrocyte / 15 kDa) is an adaptor protein that regulates death receptor-induced apoptosis and proliferation signaling by binding to FADD and relocating ERK1/2 to the cytosol, respectively. By using PEA-15-deficient mice, we examined the role of PEA-15 in TCR-dependent cytokine production in CD4(+) T cells. TCR-stimulated PEA-15-deficient CD4(+) T cells exhibited defective progression through the cell cycle associated with impaired expression of cyclin E and phosphoRb, two ERK1/2-dependent proteins of the cell cycle. Accordingly, expression of the division cycle-dependent cytokines IL-2 and IFNγ, a Th1 cytokine, was reduced in stimulated PEA-15-deficient CD4(+) T cells. This was associated with abnormal subcellular compartmentalization of activated ERK1/2 in PEA-15-deficient T cells. Furthermore, in vitro TCR-dependent differentiation of naive CD4(+) CD62L(+) PEA-15-deficient T cells was associated with a lower production of the Th2 cytokine, IL-4, whereas expression of the Th17-associated molecule IL4I1 was enhanced. Finally, a defective humoral response was shown in PEA-15-deficient mice in a model of red blood cell alloimmunization performed with Poly IC, a classical adjuvant of Th1 response in vivo. Collectively, our data suggest that PEA-15 contributes to the specification of the cytokine pattern of activated Th cells, thus highlighting a potential new target to interfere with T cell functional polarization and subsequent immune response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Lymphocyte Activation/immunology , Phosphoproteins/deficiency , T-Lymphocytes, Helper-Inducer/immunology , Animals , Apoptosis Regulatory Proteins , Blood Transfusion , Disease Models, Animal , Immunization, Passive , In Vitro Techniques , MAP Kinase Signaling System , Mice , Receptors, Antigen, T-Cell/metabolism
8.
Stem Cell Reports ; 4(3): 360-73, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25684226

ABSTRACT

Previously, we described the safety and therapeutic potential of neurospheres (NSs) derived from a human induced pluripotent stem cell (iPSC) clone, 201B7, in a spinal cord injury (SCI) mouse model. However, several safety issues concerning iPSC-based cell therapy remain unresolved. Here, we investigated another iPSC clone, 253G1, that we established by transducing OCT4, SOX2, and KLF4 into adult human dermal fibroblasts collected from the same donor who provided the 201B7 clone. The grafted 253G1-NSs survived, differentiated into three neural lineages, and promoted functional recovery accompanied by stimulated synapse formation 47 days after transplantation. However, long-term observation (for up to 103 days) revealed deteriorated motor function accompanied by tumor formation. The tumors consisted of Nestin(+) undifferentiated neural cells and exhibited activation of the OCT4 transgene. Transcriptome analysis revealed that a heightened mesenchymal transition may have contributed to the progression of tumors derived from grafted cells.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Differentiation , Cell Lineage , Cell Survival , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , Computational Biology , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Heterografts , Humans , Induced Pluripotent Stem Cells/pathology , Kruppel-Like Factor 4 , Mice , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Signal Transduction , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Stem Cell Transplantation/adverse effects , Transcriptome
9.
Neurorehabil Neural Repair ; 29(7): 677-89, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25527489

ABSTRACT

BACKGROUND: Spasticity and allodynia are major sequelae that affect the quality of life and daily activities of spinal cord injury (SCI) patients. Although rehabilitation ameliorates spasticity and allodynia, the molecular mechanisms involved in these processes remain elusive. OBJECTIVE: To investigate the molecular mechanisms by which rehabilitation ameliorates spasticity and allodynia after SCI in rats. METHODS: The expression levels of brain-derived neurotrophic factor (BDNF) and potassium-chloride cotransporter-2 (KCC2), as well as the localization of KCC2, were examined in the lumbar enlargements of untrained and treadmill-trained thoracic SCI model rats. Spasticity and allodynia were determined via behavioral and electrophysiological analyses. The effects of BDNF on spasticity, allodynia, and KCC2 activation were determined by inhibition of BDNF signaling via intrathecal administration of TrkB-IgG. The effects of SCI and training on the expression levels of functional phospholipase C-γ in the lumbar enlargement were also examined. RESULTS: Treadmill training after SCI upregulated endogenous BDNF expression and posttranslational modification of KCC2 in the lumbar enlargement significantly. There were also significant correlations between increased KCC2 expression and ameliorated spasticity and allodynia. Administration of TrkB-IgG abrogated the training-induced upregulation of KCC2 and beneficial effects on spasticity and allodynia. The expression level of functional phospholipase C-γ was reduced significantly after SCI, which may have contributed to the change in the function of BDNF, whereby it did not trigger short-term downregulation or induce long-term upregulation of KCC2 expression secondary to training. CONCLUSIONS: BDNF-mediated restoration of KCC2 expression underlies the suppression of spasticity and allodynia caused by rehabilitation.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hyperalgesia/rehabilitation , Muscle Spasticity/rehabilitation , Physical Therapy Modalities , Up-Regulation/physiology , Animals , Calcitonin Gene-Related Peptide/metabolism , Disease Models, Animal , Exercise Test , Female , Hyperalgesia/etiology , Immunoglobulin G/therapeutic use , Locomotion , Muscle Spasticity/etiology , Neurons/metabolism , Phospholipase C gamma/metabolism , Rats , Rats, Sprague-Dawley , Receptor, trkB/immunology , Spinal Cord Injuries/complications , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Statistics, Nonparametric , Symporters/metabolism , Time Factors , K Cl- Cotransporters
10.
Stem Cells Dev ; 23(18): 2250-61, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25058468

ABSTRACT

The transcriptional regulation of neural stem/progenitor cells (NS/PCs) is of great interest in neural development and stem cell biology. The RNA-binding protein Musashi1 (Msi1), which is often employed as a marker for NS/PCs, regulates Notch signaling to maintain NS/PCs in undifferentiated states by the translational repression of Numb expression. Considering these critical roles of Msi1 in the maintenance of NS/PCs, it is extremely important to elucidate the regulatory mechanisms by which Msi1 is selectively expressed in these cells. However, the mechanism regulating Msi1 transcription is unclear. We previously reported that the transcriptional regulatory region of Msi1 is located in the sixth intron of the Msi1 locus in NS/PCs, based on in vitro experiments. In the present study, we generated reporter transgenic mice for the sixth intronic Msi1 enhancer (Msi1-6IE), which show the reporter expression corresponding with endogenous Msi1-positive cells in developing and adult NS/PCs. We found that the core element responsible for this reporter gene activity includes palindromic Regulatory factor X (Rfx) binding sites and that Msi1-6IE was activated by Rfx. Rfx4, which was highly expressed in NS/PCs positive for the Msi1-6IE reporter, bound to this region, and both of the palindromic Rfx binding sites were required for the transactivation of Msi1-6IE. Furthermore, ectopic Rfx4 expression in the developing mouse cerebral cortex transactivates Msi1 expression in the intermediate zone. This study suggests that ciliogenic Rfx transcription factors regulate Msi1 expression through Msi1-6IE in NS/PCs.


Subject(s)
DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , RNA-Binding Proteins/genetics , Transcription Factors/metabolism , Animals , Base Pairing/genetics , Binding Sites , Cell Proliferation , Enhancer Elements, Genetic/genetics , Genes, Reporter , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Prosencephalon/embryology , Prosencephalon/metabolism , Protein Binding , RNA-Binding Proteins/metabolism , Regulatory Factor X Transcription Factors , Transcription, Genetic , Transcriptional Activation/genetics
11.
Stem Cell Reports ; 1(2): 152-65, 2013.
Article in English | MEDLINE | ID: mdl-24052950

ABSTRACT

Human mesenchymal stem cells (hMSCs), which conventionally are isolated based on their adherence to plastic, are heterogeneous and have poor growth and differentiation, limiting our ability to investigate their intrinsic characteristics. We report an improved prospective clonal isolation technique and reveal that the combination of three cell-surface markers (LNGFR, THY-1, and VCAM-1) allows for the selection of highly enriched clonogenic cells (one out of three isolated cells). Clonal characterization of LNGFR(+)THY-1(+) cells demonstrated cellular heterogeneity among the clones. Rapidly expanding clones (RECs) exhibited robust multilineage differentiation and self-renewal potency, whereas the other clones tended to acquire cellular senescence via P16INK4a and exhibited frequent genomic errors. Furthermore, RECs exhibited unique expression of VCAM-1 and higher cellular motility compared with the other clones. The combination marker LNGFR(+)THY-1(+)VCAM-1(hi+) (LTV) can be used selectively to isolate the most potent and genetically stable MSCs.


Subject(s)
Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Thy-1 Antigens/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Femur/cytology , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Mice , Middle Aged , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Receptors, Nerve Growth Factor/genetics , Thy-1 Antigens/genetics , Vascular Cell Adhesion Molecule-1/genetics , Young Adult
12.
EMBO Mol Med ; 3(11): 682-96, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21898827

ABSTRACT

The migratory response of astrocytes is essential for restricting inflammation and preserving tissue function after spinal cord injury (SCI), but the mechanisms involved are poorly understood. Here, we observed stimulation of in vitro astrocyte migration by the new potent glycogen synthase kinase-3 (GSK-3) inhibitor Ro3303544 and investigated the effect of Ro3303544 administration for 5 days following SCI in mice. This treatment resulted in accelerated migration of reactive astrocytes to sequester inflammatory cells that spared myelinated fibres and significantly promoted functional recovery. Moreover, the decreased extent of chondroitin sulphate proteoglycans and collagen IV demonstrated that scarring was reduced in Ro3303544-treated mice. A variety of in vitro and in vivo experiments further suggested that GSK-3 inhibition stimulated astrocyte migration by decreasing adhesive activity via reduced surface expression of ß1-integrin. Our results reveal a novel benefit of GSK-3 inhibition for SCI and suggest that the stimulation of astrocyte migration is a feasible therapeutic strategy for traumatic injury in the central nervous system.


Subject(s)
Astrocytes/cytology , Cell Movement , Down-Regulation , Glycogen Synthase Kinase 3/genetics , Spinal Cord Injuries/enzymology , Spinal Cord Injuries/physiopathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Adhesion/drug effects , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Mice , Mice, Inbred C57BL , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/genetics
13.
Mol Brain ; 3: 31, 2010 Oct 31.
Article in English | MEDLINE | ID: mdl-21034515

ABSTRACT

BACKGROUND: While several mouse strains have recently been developed for tracing neural crest or oligodendrocyte lineages, each strain has inherent limitations. The connection between human SOX10 mutations and neural crest cell pathogenesis led us to focus on the Sox10 gene, which is critical for neural crest development. We generated Sox10-Venus BAC transgenic mice to monitor Sox10 expression in both normal development and in pathological processes. RESULTS: Tissue fluorescence distinguished neural crest progeny cells and oligodendrocytes in the Sox10-Venus mouse embryo. Immunohistochemical analysis confirmed that Venus expression was restricted to cells expressing endogenous Sox10. Time-lapse imaging of various tissues in Sox10-Venus mice demonstrated that Venus expression could be visualized at the single-cell level in vivo due to the intense, focused Venus fluorescence. In the adult Sox10-Venus mouse, several types of mature and immature oligodendrocytes along with Schwann cells were clearly labeled with Venus, both before and after spinal cord injury. CONCLUSIONS: In the newly-developed Sox10-Venus transgenic mouse, Venus fluorescence faithfully mirrors endogenous Sox10 expression and allows for in vivo imaging of live cells at the single-cell level. This Sox10-Venus mouse will thus be a useful tool for studying neural crest cells or oligodendrocytes, both in development and in pathological processes.


Subject(s)
Bacterial Proteins/metabolism , Luminescent Proteins/metabolism , Mice, Transgenic , Neural Crest/cytology , Oligodendroglia/metabolism , SOXE Transcription Factors/metabolism , Staining and Labeling/methods , Time-Lapse Imaging/methods , Animals , Bacterial Proteins/genetics , Cell Lineage , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Female , Genes, Reporter , Humans , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Oligodendroglia/cytology , SOXE Transcription Factors/genetics , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
14.
Exp Neurol ; 224(2): 403-14, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20478301

ABSTRACT

We previously reported the beneficial effect of administering an anti-mouse IL-6 receptor antibody (MR16-1) immediately after spinal cord injury (SCI). The purpose of our present study was to clarify the mechanism underlying how MR16-1 improves motor function after SCI. Quantitative analyses of inflammatory cells using flow cytometry, and immunohistochemistry with bone marrow-chimeric mice generated by transplanting genetically marked purified hematopoietic stem cells, revealed that MR16-1 dramatically switched the central player in the post-traumatic inflammation, from hematogenous macrophages to resident microglia. This change was accompanied by alterations in the expression of relevant cytokines within the injured spinal cord; the expression of recruiting chemokines including CCL2, CCL5, and CXCL10 was decreased, while that of Granulocyte/Macrophage-Colony Stimulating Factor (GM-CSF), a known mitogen for microglia, was increased. We also showed that the resident microglia expressed higher levels of phagocytic markers than the hematogenous macrophages. Consistent with these findings, we observed significantly decreased tissue damage and reduced levels of myelin debris and Nogo-A, the axonal growth inhibitor, by MR16-1 treatment. Moreover, we observed increased axonal regeneration and/or sprouting in the MR16-1-treated mice. Our findings indicate that the functional improvement elicited by MR16-1 involves microglial functions, and provide new insights into the role of IL-6 signaling in the pathology of SCI.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Microglia/drug effects , Receptors, Interleukin-6/immunology , Spinal Cord Injuries/drug therapy , Animals , Chemokines/biosynthesis , Chimera , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cell Transplantation , Inflammation/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Transgenic , Microglia/immunology , Phagocytosis , Rats , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology
15.
BMC Cancer ; 10: 66, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20181261

ABSTRACT

BACKGROUND: Tumor initiating cells (TICs) provide a new paradigm for developing original therapeutic strategies. METHODS: We screened for TICs in 47 human adult brain malignant tumors. Cells forming floating spheres in culture, and endowed with all of the features expected from tumor cells with stem-like properties were obtained from glioblastomas, medulloblastoma but not oligodendrogliomas. RESULTS: A long-term self-renewal capacity was particularly observed for cells of malignant glio-neuronal tumors (MGNTs). Cell sorting, karyotyping and proteomic analysis demonstrated cell stability throughout prolonged passages. Xenografts of fewer than 500 cells in Nude mouse brains induced a progressively growing tumor. CD133, CD15/LeX/Ssea-1, CD34 expressions, or exclusion of Hoechst dye occurred in subsets of cells forming spheres, but was not predictive of their capacity to form secondary spheres or tumors, or to resist high doses of temozolomide. CONCLUSIONS: Our results further highlight the specificity of a subset of high-grade gliomas, MGNT. TICs derived from these tumors represent a new tool to screen for innovative therapies.


Subject(s)
Antigens, CD34/biosynthesis , Antigens, CD/biosynthesis , Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Glycoproteins/biosynthesis , Lewis X Antigen/biosynthesis , Neoplastic Stem Cells/cytology , Neurons/pathology , AC133 Antigen , Animals , Cell Line, Tumor , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Peptides , Proteomics/methods
16.
Int J Biochem Cell Biol ; 40(9): 1649-53, 2008.
Article in English | MEDLINE | ID: mdl-18434236

ABSTRACT

Spinal cord injury (SCI), despite considerable progress in palliative care, has currently no satisfying therapeutic leading to functional recovery. Inability of central nervous system severed axons to regenerate after injury is considered to originate from both limited intrinsic capabilities of neurons and inhibitory effect of the local environment. Precisely, the so-called "glial scar" formed by reactive astrocytes in response to injury exerts a well-known axon-outgrowth inhibitory effect. However, recent studies revealed that role of reactive astrocytes after SCI is more complex. During the first weeks after injury, reactive astrocytes indeed protect the tissue and contribute to a spontaneous relative functional recovery. Compaction of the lesion center and seclusion of inflammatory cells by migrating reactive astrocytes seem to underlie this beneficial effect. Stimulation of reactive astrocytes migration in the sub-acute phase of SCI might thus represent a new approach to improve the functional outcome of patients.


Subject(s)
Astrocytes/pathology , Cell Movement , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology , Animals , Astrocytes/cytology , Humans , Inflammation/complications , Inflammation/pathology
17.
Mol Biol Cell ; 17(12): 5141-52, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16987961

ABSTRACT

Phosphoprotein enriched in astrocytes-15 kDa (PEA-15), a phosphoprotein enriched in astrocytes, inhibits both apoptosis and proliferation in normal and cancerous cells. Here, analysis of PEA-15 expression in glioblastoma organotypic cultures revealed low levels of PEA-15 in tumor cells migrating away from the explants, regardless of the expression levels in the originating explants. Because glioblastomas are highly invasive primary brain tumors that can originate from astrocytes, we explored the involvement of PEA-15 in the control of astrocyte migration. PEA-15-/- astrocytes presented an enhanced motility in vitro compared with their wild-type counterparts. Accordingly, NIH-3T3 cells transfected by green fluorescent protein-PEA-15 displayed a reduced migration. Reexpression of PEA-15 restored PEA-15-/- astrocyte motility to wild-type levels. Pharmacological manipulations excluded a participation of extracellular signal-regulated kinase/mitogen-activated protein kinase, phosphatidylinositol 3-kinase/Akt, and calcium/calmodulin-dependent protein kinase II in this effect of PEA-15. In contrast, treatment by bisindolylmaleimide, Gö6976, and rottlerin, and chronic application of phorbol 12-myristate 13-acetate and/or bryostatin-1 indicated that PKC delta mediated PEA-15 inhibition of astrocyte migration. PEA-15-/- astrocytes constitutively expressed a 40-kDa form of PKC delta that was down-regulated upon PEA-15 reexpression. Together, these data reveal a new function for PEA-15 in the inhibitory control of astrocyte motility through a PKC delta-dependent pathway involving the constitutive expression of a catalytic fragment of PKC delta.


Subject(s)
Astrocytes/cytology , Cell Movement , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Protein Kinase C-delta/metabolism , Active Transport, Cell Nucleus , Animals , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioblastoma/pathology , Humans , Mice , Molecular Weight , NIH 3T3 Cells , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...