Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Lett ; 335(1): 201-4, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23419525

ABSTRACT

Exosomes are 30-150 nm vesicles secreted by a wide range of mammalian cells that can contain microRNA (miRNA). To test if marrow stromal cell (MSC) exosomes could be used as a vehicle for delivery of anti-tumor miRNAs, we transfected MSCs with a miR-146b expression plasmid, and harvested exosomes released by the MSCs. Intra-tumor injection of exosomes derived from miR-146-expressing MSCs significantly reduced glioma xenograft growth in a rat model of primary brain tumor.


Subject(s)
Brain Neoplasms/therapy , Exosomes/metabolism , Glioma/therapy , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Animals , Brain Neoplasms/pathology , Gene Transfer Techniques , Genetic Therapy , Glioma/pathology , Humans , Injections, Intralesional , Male , MicroRNAs/administration & dosage , MicroRNAs/genetics , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Tumor Burden
2.
Stem Cells ; 30(7): 1556-64, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22605481

ABSTRACT

Multipotent mesenchymal stromal cells (MSCs) have potential therapeutic benefit for the treatment of neurological diseases and injury. MSCs interact with and alter brain parenchymal cells by direct cell-cell communication and/or by indirect secretion of factors and thereby promote functional recovery. In this study, we found that MSC treatment of rats subjected to middle cerebral artery occlusion (MCAo) significantly increased microRNA 133b (miR-133b) level in the ipsilateral hemisphere. In vitro, miR-133b levels in MSCs and in their exosomes increased after MSCs were exposed to ipsilateral ischemic tissue extracts from rats subjected to MCAo. miR-133b levels were also increased in primary cultured neurons and astrocytes treated with the exosome-enriched fractions released from these MSCs. Knockdown of miR-133b in MSCs confirmed that the increased miR-133b level in astrocytes is attributed to their transfer from MSCs. Further verification of this exosome-mediated intercellular communication was performed using a cel-miR-67 luciferase reporter system and an MSC-astrocyte coculture model. Cel-miR-67 in MSCs was transferred to astrocytes via exosomes between 50 and 100 nm in diameter. Our data suggest that the cel-miR-67 released from MSCs was primarily contained in exosomes. A gap junction intercellular communication inhibitor arrested the exosomal microRNA communication by inhibiting exosome release. Cultured neurons treated with exosome-enriched fractions from MSCs exposed to 72 hours post-MCAo brain extracts significantly increased the neurite branch number and total neurite length. This study provides the first demonstration that MSCs communicate with brain parenchymal cells and may regulate neurite outgrowth by transfer of miR-133b to neural cells via exosomes.


Subject(s)
Exosomes/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Neurites/metabolism , Neurogenesis/physiology , Neurons/cytology , Neurons/metabolism , Animals , Cells, Cultured , Exosomes/ultrastructure , Male , Mesenchymal Stem Cells/ultrastructure , Microscopy, Electron, Transmission , Neurites/ultrastructure , Neurogenesis/genetics , Neurons/ultrastructure , Rats , Rats, Wistar
3.
Neurobiol Dis ; 46(1): 157-64, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22266016

ABSTRACT

BACKGROUND AND OBJECTIVE: We investigated axonal plasticity in the bilateral motor cortices and the long term therapeutic effect of Niaspan on axonal remodeling after stroke in type-1 diabetic (T1DM) rats. EXPERIMENTAL APPROACHES: T1DM was induced in young adult male Wistar rats via injection of streptozotocin. T1DM rats were subjected to 2h transient middle cerebral artery occlusion (MCAo) and were treated with 40 mg/kg Niaspan or saline starting 24 h after MCAo and daily for 28 days. Anterograde tracing using biotinylated dextran amine (BDA) injected into the contralateral motor cortex was performed to assess axonal sprouting in the ipsilateral motor cortex area. Functional outcome, SMI-31 (a pan-axonal microfilament marker), Bielschowsky silver and synaptophysin expression were measured. In vitro studies using primary cortical neuron (PCN) cultures and in vivo BDA injection into the brain to anterogradely label axons and terminals were employed. RESULTS: Niaspan treatment of stroke in T1DM-MCAo rats significantly improved functional outcome after stroke and increased SMI-31, Bielschowsky silver and synaptophysin expression in the ischemic brain compared to saline treated T1DM-MCAo rats (p<0.05). Using BDA to anterograde label axons and terminals, Niaspan treatment significantly increased axonal density in ipsilateral motor cortex in T1DM-MCAo rats (p<0.05, n=7/group). Niacin treatment of PCN significantly increased Ang1 expression under high glucose condition. Niacin and Ang1 significantly increased neurite outgrowth, and anti-Ang1 antibody marginally attenuated Niacin induced neurite outgrowth (p=0.06, n=6/group) in cultured PCN under high glucose condition. CONCLUSION: Niaspan treatment increased ischemic brain Ang1 expression and promoted axonal remodeling in the ischemic brain as well as improved functional outcome after stroke. Ang1 may partially contribute to Niaspan-induced axonal remodeling after stroke in T1DM-rats.


Subject(s)
Axons/drug effects , Diabetes Mellitus, Type 1/complications , Infarction, Middle Cerebral Artery/drug therapy , Neuronal Plasticity/drug effects , Niacin/pharmacology , Animals , Axons/physiology , Diabetes Mellitus, Type 1/physiopathology , Disease Models, Animal , Female , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Male , Neuronal Plasticity/physiology , Niacin/analogs & derivatives , Pregnancy , Primary Cell Culture , Rats , Rats, Wistar , Vitamin B Complex/pharmacology
4.
Cancer Sci ; 102(7): 1350-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21477071

ABSTRACT

Analysis of microarray probe data from glioma patient samples, in conjunction with patient Kaplan-Meier survival plots, indicates that expression of a glioma suppressor gene doublecortin (DCX) favors glioma patient survival. From neurosphere formation in culture, time-lapse microscopic video recording, and tumor xenograft, we show that DCX synthesis significantly reduces self-renewal of brain tumor stem cells (BTSC) in human primary glioma (YU-PG, HF66) cells from surgically removed human glioma specimens and U87 cells in vitro and in vivo. Time-lapse microscopic video recording revealed that double transfection of YU-PG, HF66, and U87 cells with DCX and neurabin II caused incomplete cell cycle with failure of cytokinesis, that is, endomitosis by dividing into three daughter cells from one mother BTSC. Activation of c-jun NH2-terminal kinase 1 (JNK1) after simvastatin (10 nM) treatment of DCX(+) neurabin II(+) BTSC from YU-PG, HF66, and U87 cells induced terminal differentiation into neuron-like cells. dUTP nick end labeling data indicated that JNK1 activation also induced apoptosis only in double transfected BTSC with DCX and neurabin II, but not in single transfected BTSC from YU-PG, HF66, and U87 cells. Western blot analysis showed that procaspase-3 was induced after DCX transfection and activated after simvastatin treatment in YU-PG, HF66, and U87 BTSC. Sequential immunoprecipitation and Western blot data revealed that DCX synthesis blocked protein phosphatase-1 (PP1)/caspase-3 protein-protein interaction and increased PP1-DCX interaction. These data show that DCX synthesis induces apoptosis in BTSC through a novel JNK1/neurabin II/DCX/PP1/caspase-3 pathway.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Microtubule-Associated Proteins/physiology , Neoplastic Stem Cells/pathology , Neuropeptides/physiology , Animals , Apoptosis/drug effects , Brain Neoplasms/mortality , Caspase 3/physiology , Cell Differentiation , Cell Line, Tumor , Doublecortin Domain Proteins , Doublecortin Protein , Glioma/mortality , Humans , Male , Microfilament Proteins/physiology , Mitogen-Activated Protein Kinase 8/physiology , Nerve Tissue Proteins/physiology , Protein Phosphatase 1/physiology , Rats , Simvastatin/pharmacology
5.
FEBS J ; 277(20): 4299-307, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20840605

ABSTRACT

MicroRNAs are small RNAs that attenuate protein expression by complementary binding to the 3'-UTR of a target mRNA. Currently, very little is known about microRNAs after cerebral ischemia. In particular, microRNA-21 (miR-21) is a strong antiapoptotic factor in some biological systems. We investigated the role of miR-21 after stroke in the rat. We employed in situ hybridization and laser capture microdissection in combination with real-time RT-PCR to investigate the expression of miR-21 after stroke. In situ hybridization revealed that miR-21 expression was upregulated in neurons of the ischemic boundary zone, and quantitative real-time RT-PCR analysis revealed that stroke increased mature miR-21 levels by approximately threefold in neurons isolated from the ischemic boundary zone by laser capture microdissection as compared with homologous contralateral neurons 2 days (n = 4; P < 0.05) and 7 days (n = 3; P < 0.05) after stroke. In vitro, overexpression of miR-21 in cultured cortical neurons substantially suppressed oxygen and glucose deprivation-induced apoptotic cell death, whereas attenuation of endogenous miR-21 by antisense inhibition exacerbated cell death after oxygen and glucose deprivation. Moreover, overexpression of miR-21 in neurons significantly reduced FASLG levels, and introduction of an miR-21 mimic into 293-HEK cells substantially reduced luciferase activity in a reporter system containing the 3'-UTR of Faslg. Our data indicate that overexpression of miR-21 protects against ischemic neuronal death, and that downregulation of FASLG, a tumor necrosis factor-α family member and an important cell death-inducing ligand whose gene is targeted by miR-21, probably mediates the neuroprotective effect. These novel findings suggest that miR-21 may be an attractive therapeutic molecule for treatment of stroke.


Subject(s)
Fas Ligand Protein/physiology , Ischemia/pathology , MicroRNAs/physiology , Neurons/pathology , Animals , Cell Death , Fas Ligand Protein/genetics , Gene Expression Regulation , MicroRNAs/analysis , MicroRNAs/genetics , Protective Agents , Rats , Rats, Wistar , Stroke
6.
Stroke ; 41(5): 1001-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20203318

ABSTRACT

BACKGROUND AND PURPOSE: Treatment with a selective proteasome inhibitor, VELCADE, in combination with tissue plasminogen activator (tPA) extended the therapeutic window to 6 hours in young rats after stroke. However, stroke is a major cause of death and disability in the elderly. The present study investigated the effect of VELCADE in combination with a low-dose tPA on aged rats after embolic stroke. METHODS: Male Wistar rats at the age of 18 to 20 months were treated with VELCADE (0.2 mg/kg) alone, a low-dose tPA (5 mg/kg) alone, combination of VELCADE and tPA, or saline 2 hours after embolic middle cerebral artery occlusion. To test the contribution of endothelial nitric oxide synthase to VELCADE-mediated neuroprotection, endothelial nitric oxide synthase knockout and wild-type mice were treated with VELCADE (0.5 mg/kg) 2 hours after embolic stroke. RESULTS: Treatment with VELCADE significantly reduced infarct volume, whereas tPA alone did not reduce infarct volume and aggravated blood-brain barrier disruption in aged rats compared with saline-treated rats. However, the combination treatment significantly enhanced the reduction of infarct volume, which was associated with an increase in endothelial nitric oxide synthase activity compared with saline-treated rats. Additionally, the combination treatment promoted thrombolysis and did not increase the incidence of hemorrhage transformation. VELCADE significantly reduced lesion volume in wild-type mice but failed to significantly reduce lesion volume in endothelial nitric oxide synthase knockout mice. CONCLUSIONS: Treatment with VELCADE exerts a neuroprotective effect in aged rats after stroke. The combination of VELCADE with the low-dose tPA further amplifies the neuroprotective effect. Endothelial nitric oxide synthase at least partly contributes to VELCADE-mediated neuroprotection after stroke.


Subject(s)
Aging/drug effects , Boronic Acids/administration & dosage , Brain Ischemia/prevention & control , Intracranial Embolism/prevention & control , Neuroprotective Agents/administration & dosage , Pyrazines/administration & dosage , Tissue Plasminogen Activator/administration & dosage , Aging/pathology , Animals , Bortezomib , Brain Ischemia/enzymology , Brain Ischemia/pathology , Drug Administration Schedule , Drug Therapy, Combination , Intracranial Embolism/enzymology , Intracranial Embolism/pathology , Male , Protease Inhibitors/administration & dosage , Rats , Rats, Wistar
7.
J Biol Chem ; 284(34): 22680-9, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19553662

ABSTRACT

Ischemic stroke stimulates neurogenesis in the adult rodent brain. The molecules underlying stroke-induced neurogenesis have not been fully investigated. Using real-time reverse transcription-PCR, we found that stroke substantially up-regulated angiopoietin 2 (ANG2), a proangiogenic gene, expression in subventricular zone neural progenitor cells. Incubation of neural progenitor cells with recombinant human ANG2 significantly increased the number of beta-III tubulin-positive cells, a marker of immature neurons, but did not alter the number of glial fibrillary acidic protein (GFAP)-positive cells, a marker of astrocytes, suggesting that ANG2 promotes neuronal differentiation. Blockage of the ANG2 receptor, Tie2, with small interference RNA (siRNA)-Tie2 attenuated recombinant human ANG2 (rhANG2)-increased beta-III tubulin mRNA levels compared with levels in the progenitor cells transfected with control siRNA. Chromatin immunoprecipitation analysis revealed that CCAAT/enhancer-binding protein (C/EBP beta) up-regulated by rhANG2 bound to beta-III tubulin, which is consistent with published data that there are several C/EBP beta binding sites in the promoter of beta-III tubulin gene. In addition, rhANG2 enhanced migration of neural progenitor cells measured by single neurosphere assay. Blockage of Tie2 with siRNA-Tie2 and a Tie2-neutralizing antibody did not suppress ANG2-enhanced migration. However, inhibition of matrix metalloproteinases with GM6001 blocked ANG2-enhanced migration. Collectively, our data suggest that interaction of ANG2, a proangiogenic factor, with its receptor Tie2 promotes neural progenitor cell differentiation into neuronal lineage cells, whereas ANG2 regulates neural progenitor cell migration through matrix metalloproteinases, which do not require its receptor Tie2.


Subject(s)
Angiopoietin-2/pharmacology , Angiopoietin-2/physiology , Cell Differentiation/drug effects , Cell Movement/drug effects , Neurons/metabolism , Stem Cells/metabolism , Stroke/physiopathology , Angiopoietin-2/genetics , Animals , Brain/cytology , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Cells, Cultured , Chromatin Immunoprecipitation , Dipeptides/pharmacology , Electroporation , Gene Expression/drug effects , Humans , Immunohistochemistry , In Vitro Techniques , Male , Metalloendopeptidases/antagonists & inhibitors , Mice , Microdissection , Neurons/cytology , Neurons/drug effects , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Receptor, TIE-2/physiology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/drug effects
8.
J Cereb Blood Flow Metab ; 29(6): 1166-74, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19384336

ABSTRACT

Oligodendrocytes are sensitive to ischemic damage. The Sonic hedgehog (Shh) pathway is critical in oligodendrogenesis; Gli1 is the principal effector of Shh signaling. We investigated oligodendrogenesis and Shh/Gli1 pathway activation after bone marrow stromal cell (BMSC) treatment of stroke in rats. Rats were subjected to the middle cerebral artery occlusion (MCAo). BMSCs have been shown to promote functional recovery post stroke. A therapeutic dose of BMSC (3 x 10(6) cells) treatment was initiated 1 day after MCAo. Immunohistochemistry was carried out to measure the oligodendrocyte progenitor cells, oligodendrocytes, myelin, and expressions of Shh and Gli1 at 14 days after MCAo. Gene expression of Shh and Gli1 was tested at 2 days after MCAo. An in vitro study was used to investigate the effects of BMSC on a premature oligodendrocyte cell line (N20.1 cells). BMSC treatment significantly increased O4(+) oligodendrocytes, MBP(+) area, and bromodeoxyuridine (BrdU)(+), NG2(+), BrdU(+)-NG2(+) cells, and mRNA and protein expressions of Shh and Gli1 in the ipsilateral brain of the MCAo rats than that in phosphate buffered saline (PBS)-treated rats. BMSCs promoted N20.1 cell proliferation and Gli1 mRNA expression, and these effects were abolished by the Shh pathway inhibitor cyclopamine. These data indicate that the BMSC treatment stimulates oligodendrogenesis by activation of the Shh/Gli1 pathway post stroke.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation , Oligodendroglia/cytology , Stroke/pathology , Animals , Bone Marrow Cells/metabolism , Cell Proliferation , Cells, Cultured , Hedgehog Proteins/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Mice , Myelin Sheath , Oncogene Proteins/metabolism , Rats , Rats, Wistar , Signal Transduction , Stroke/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Zinc Finger Protein GLI1
9.
J Cereb Blood Flow Metab ; 29(2): 297-307, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18854839

ABSTRACT

Stroke increases neuroblasts in the subventricular zone (SVZ) of the lateral ventricle and these neuroblasts migrate toward the ischemic boundary to replace damaged neurons. Using brain slices from the nonischemic adult rat and transgenic mice that expressed enhanced green fluorescent protein (EGFP) concomitantly with doublecortin (DCX), a marker for migrating neuroblasts, we recorded electrophysiological characteristics while simultaneously analyzing the gene expression in single SVZ cells. We found that SVZ cells expressing the DCX gene from the nonischemic rat had a mean resting membrane potential (RMP) of -30 mV. DCX-EGFP-positive cells in the nonischemic SVZ of the transgenic mouse had a mean RMP of -25+/-7 mV and did not exhibit Na(+) currents, characteristic of immature neurons. However, DCX-EGFP-positive cells in the ischemic SVZ exhibited a hyperpolarized mean RMP of -54+/-18 mV and displayed Na(+) currents, indicative of more mature neurons. Single-cell multiplex RT-PCR analysis revealed that DCX-EGFP-positive cells in the nonischemic SVZ of the transgenic mouse expressed high neural progenitor marker genes, Sox2 and nestin, but not mature neuronal marker genes. In contrast, DCX-EGFP-positive cells in the ischemic SVZ expressed tyrosine hydroxylase, a mature neuronal marker gene. Together, these data indicate that stroke changes gene profiles and the electrophysiology of migrating neuroblasts.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation/genetics , Membrane Potentials , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Stroke/metabolism , Animals , Doublecortin Domain Proteins , Doublecortin Protein , Electrophysiology , Female , Male , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Neuropeptides/genetics , Patch-Clamp Techniques , Rats , Stroke/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...