Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
RNA Biol ; 14(5): 488-499, 2017 05 04.
Article in English | MEDLINE | ID: mdl-27315491

ABSTRACT

Neurogenesis is associated with functional recovery after stroke. However, the underlying molecular mechanisms have not been fully investigated. Using an Ago2-based RNA immunoprecipitation to immunoprecipated Ago2-RNA complexes followed by RNA sequencing (Ago2 RIP-seq) approach, we profiled the miRNomes in neural progenitor cells (NPCs) harvested from the subventricular zone (SVZ) of the lateral ventricles of young adult rats. We identified more than 7 and 15 million reads in normal and ischemic NPC libraries, respectively. We found that stroke substantially changed Ago2-associated miRNA profiles in NPCs compared to those in non-ischemic NPCs. We also discovered a new complex repertoire of isomiRs and multiple miRNA-miRNA* pairs and numerous novel miRNAs in the non-ischemic and ischemic NPCs. Among them, pc-3p-17172 significantly regulated NPC proliferation and neuronal differentiation. Collectively, the present study reveals profiles of Ago2-associated miRNomes in non-ischemic and ischemic NPCs, which provide a molecular basis to further investigate the role of miRNAs in mediating adult neurogenesis under physiological and ischemic conditions.


Subject(s)
Argonaute Proteins/metabolism , MicroRNAs/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Stroke/metabolism , Adult , Analysis of Variance , Animals , Argonaute Proteins/genetics , Cell Proliferation , Humans , Lateral Ventricles/chemistry , Male , MicroRNAs/analysis , MicroRNAs/genetics , Neural Stem Cells/pathology , Primary Cell Culture , Rats , Rats, Wistar , Sequence Analysis, RNA , Stroke/pathology , Transcriptome
2.
J Biol Chem ; 288(18): 12478-88, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23511639

ABSTRACT

The role of microRNAs (miRNAs) in mediating adult neurogenesis after stroke has not been extensively studied. The present study investigated the function of the miR17-92 cluster in adult neural progenitor cells after experimental stroke. We found that stroke substantially up-regulated miR17-92 cluster expression in neural progenitor cells of the adult mouse. Overexpression of the miR17-92 cluster either in cultured ischemic neural progenitor cells or in the subventricular zone (SVZ) of ischemic animals significantly increased cell proliferation, whereas inhibition of individual members of the miR17-92 cluster, miR-18a and miR-19a, suppressed cell proliferation and increased cell death. The miR17-92 cluster mediated PTEN (phosphatase and tensin homolog) expression, which is a predicted target of the miR17-92 cluster. Addition of Sonic hedgehog (Shh) protein up-regulated miR17-92 expression and elevated c-Myc protein in ischemic neural progenitor cells, whereas blockade of the Shh signaling pathway down-regulated miR17-92 cluster expression and reduced c-Myc levels. Overexpression of c-Myc up-regulated miR17-92 cluster expression. Intraventricular infusion of Shh and a Shh receptor inhibitor, cyclopamine, to ischemic animals further elevated and suppressed, respectively, miR17-92 cluster expression in the SVZ. These data indicate that the miR17-92 cluster plays an important role in mediating neural progenitor cell function and that the Shh signaling pathway is involved in up-regulating miR17-92 cluster expression.


Subject(s)
Cell Proliferation , Gene Expression Regulation , MicroRNAs/biosynthesis , Multigene Family , Neural Stem Cells/metabolism , Signal Transduction , Animals , Cell Survival , Disease Models, Animal , Hedgehog Proteins/metabolism , Male , Mice , Nerve Tissue Proteins/metabolism , Neural Stem Cells/pathology , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-myc/metabolism
3.
Glia ; 60(12): 1906-14, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22907787

ABSTRACT

Serum response factor (SRF) is a transcription factor that transactivates actin-associated genes and has been implicated in oligodendrocyte (OL) differentiation. To date, it has not been investigated in cerebral ischemia. We investigated the dynamics of SRF expression after stroke in vivo and the role of SRF in OL differentiation in vitro. Using immunohistochemistry, we found that SRF was upregulated in OLs and OL precursor cells (OPCs) after stroke. Moreover, upregulation of SRF was concurrent with downregulation of the micro-RNAs (miRNAs) miR-9 and the miR-200 family in the ischemic white matter region, the corpus callosum. Inhibition of SRF activation by CCG-1423, a specific inhibitor of SRF function, blocked OPCs from differentiating into OLs. Overexpression of miR-9 and miR-200 in cultured OPCs suppressed SRF expression and inhibited OPC differentiation. Moreover, co-expression of miR-9 and miR-200 attenuated activity of a luciferase reporter assay containing the Srf 3' untranslated region. Collectively, this study is the first to show that stroke upregulates SRF expression in OPCs and OLs, and that SRF levels are mediated by miRNAs and regulate OPC differentiation.


Subject(s)
Cell Differentiation/physiology , MicroRNAs/physiology , Oligodendroglia/physiology , Serum Response Factor/biosynthesis , Stem Cells/physiology , Animals , Cells, Cultured , Male , MicroRNAs/antagonists & inhibitors , Oligodendroglia/cytology , Rats , Rats, Wistar , Serum Response Factor/physiology , Stem Cells/cytology , Stroke/metabolism , Stroke/pathology
4.
Neurobiol Dis ; 48(3): 546-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22922221

ABSTRACT

Peripheral neuropathy is one of the most common complications of diabetes mellitus. Using a mouse model of diabetic peripheral neuropathy, we tested the hypothesis that thymosin ß4 (Tß4) ameliorates diabetes-induced neurovascular dysfunction in the sciatic nerve and promotes recovery of neurological function from diabetic peripheral neuropathy. Tß4 treatment of diabetic mice increased functional vascular density and regional blood flow in the sciatic nerve, and improved nerve function. Tß4 upregulated angiopoietin-1 (Ang1) expression, but suppressed Ang2 expression in endothelial and Schwann cells in the diabetic sciatic nerve. In vitro, incubation of Human Umbilical Vein Endothelial Cells (HUVECs) with Tß4 under high glucose condition completely abolished high glucose-downregulated Ang1 expression and high glucose-reduced capillary-like tube formation. Moreover, incubation of HUVECs under high glucose with conditioned medium collected from Human Schwann Cells (HSCs) treated with Tß4 significantly reversed high glucose-decreased capillary-like tube formation. PI3K/Akt signaling pathway is involved in Tß4-regulated Ang1 expression on endothelial and Schwann cells. These data indicate that Tß4 likely acts on endothelial cells and Schwann cells to preserve and/or restore vascular function in the sciatic nerve which facilitates improvement of peripheral nerve function under diabetic neuropathy. Thus, Tß4 has potential for the treatment of diabetic peripheral neuropathy.


Subject(s)
Diabetic Neuropathies/metabolism , Neuroprotective Agents/pharmacology , Sciatic Nerve/drug effects , Thymosin/pharmacology , Animals , Blotting, Western , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetic Neuropathies/physiopathology , Disease Models, Animal , Electrophysiology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Immunohistochemistry , Laser-Doppler Flowmetry , Mice , Real-Time Polymerase Chain Reaction , Regional Blood Flow , Reverse Transcriptase Polymerase Chain Reaction , Schwann Cells/drug effects , Schwann Cells/metabolism , Sciatic Nerve/blood supply , Sciatic Nerve/metabolism , Signal Transduction/drug effects
5.
Stroke ; 42(3): 740-4, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21307396

ABSTRACT

BACKGROUND AND PURPOSE: We tested the effect of bone marrow stromal cells (BMSCs) on neuronal remodeling of the corticospinal tract originating from the contralesional cortex in mice subjected to unilateral pyramidotomy (PT) followed by middle cerebral artery occlusion (MCAO). METHODS: Adult mice with transgenic yellow fluorescent protein labeling in the corticospinal tract were subjected to right hemispheric PT and right permanent or sham MCAO. One day later, the mice were treated intravenously with BMSCs or phosphate-buffered saline. A Foot-Fault test and a single pellet-reaching test were performed before surgery, 3 days after MCAO, and weekly thereafter. Pseudorabies virus-614-monomeric red fluorescent protein was injected into the left forelimb flexor muscles 28 days after surgery (4 days before euthanasia). The brain and cervical cord were processed for fluorescent microscopy to detect red fluorescent protein and yellow fluorescent protein labeling, respectively. RESULTS: Significant functional improvements were evident in PT-MCAO mice treated with BMSCs (n=9) compared with phosphate-buffered saline-treated mice (n=9, P<0.05), but not in mice with PT-sham MCAO treated with either phosphate-buffered saline (n=9) or BMSCs (n=10). Furthermore, in PT-MCAO mice, both corticospinal tract axonal density in the denervated side of the cervical gray matter and red fluorescent protein-labeled pyramidal neurons in the left intact cortex were significantly increased compared with PT-sham MCAO mice (P<0.05). BMSCs significantly enhanced both corticospinal tract density and red fluorescent protein labeling in PT-MCAO mice (P<0.05) only. The behavioral outcome was highly correlated with corticospinal tract density and red fluorescent protein labeling. CONCLUSIONS: BMSCs amplify stroke-induced contralesional neuronal remodeling, which contributes to motor recovery after stroke.


Subject(s)
Axons/physiology , Bone Marrow Transplantation , Brain Ischemia/physiopathology , Motor Skills/physiology , Recovery of Function/physiology , Stroke/physiopathology , Age Factors , Animals , Brain Ischemia/complications , Brain Ischemia/surgery , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Cortex/physiology , Pyramidal Tracts/physiology , Stroke/complications , Stroke/surgery , Stromal Cells/transplantation
6.
Stroke ; 42(2): 459-64, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21212396

ABSTRACT

BACKGROUND AND PURPOSE: Bone marrow stromal cells (BMSC) decrease neurological deficits in rodents after stroke and concomitantly induce extensive neurite remodeling in the brain, which highly correlates with the improvement of neurological function. We investigated the effects of endogenous tissue plasminogen activator (tPA) on neurite remodeling after BMSC treatment. METHODS: Adult C57BL/6 wild-type (WT) mice and tPA knockout (tPA(-/-)) mice were subjected to middle cerebral artery occlusion, followed by an injection of 1×10(6) BMSC (n=18) or phosphate-buffered saline (n=18) into the tail vein 24 hours later. Behavioral tests were performed at 3, 7, and 14 days after middle cerebral artery occlusion. Animals were euthanized at 14 days after stroke. RESULTS: The effects of BMSC on functional recovery depended on presence or absence of tPA, even after adjusting for imbalanced stroke severity. BMSC significantly improve functional recovery in WT mice compared to WT controls but show no beneficial effect in the tPA(-/-) mice compared to tPA(-/-) controls. Axonal density and synaptophysin-positive areas along the ischemic boundary zone of the cortex and striatum in WT mice are significantly higher than in the tPA(-/-) mice. BMSC treatment significantly increases tPA protein level and activity only in WT mice. CONCLUSIONS: Our results suggest that endogenous tPA promotes BMSC-induced neurite outgrowth and may contribute to functional recovery after stroke.


Subject(s)
Bone Marrow Transplantation , Neurites/physiology , Stroke/surgery , Tissue Plasminogen Activator/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Bone Marrow Transplantation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Recovery of Function/physiology , Stroke/pathology , Stromal Cells/physiology
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.
Stroke ; 40(7): 2546-51, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19478220

ABSTRACT

BACKGROUND AND PURPOSE: To elucidate how the motor pathways rewire the denervated tissue after stroke, we investigated remodeling of the corticospinal tract (CST) in transgenic mice with yellow fluorescent protein CST labeling in conjunction with transsynaptic pseudorabies virus retrograde tracing. METHODS: Adult male CST-yellow fluorescent protein mice were subjected to permanent right middle cerebral artery occlusion (n=8/group). Foot-fault test was performed to monitor functional deficit and recovery. Pseudorabies virus tracer was injected into the left forelimb muscles at 1 or 4 weeks after middle cerebral artery occlusion (4 days before euthanasia), respectively. A third group of CST-yellow fluorescent protein mice without middle cerebral artery occlusion was used for normal control (n=6). The yellow fluorescent protein labeling of CST in the cervical cord and pseudorabies virus labeling of pyramidal neurons in the bilateral cortices were measured on vibratome sections using a confocal imaging system. RESULTS: Compared with normal animals, axonal density in the stroke-affected side of the cervical cord was significantly decreased at 11 days (P<0.001) and significantly increased at 32 days after stroke compared with the Day 11 values (P<0.05). Pseudorabies virus labeling was significantly decreased in the ischemic hemisphere 11 days after middle cerebral artery occlusion (P<0.001). In contrast, a significant increase was observed in pseudorabies virus labeling of bilateral cortices 32 days after stroke compared with 11 days (P<0.05). The CST axonal density in the denervated spinal cord and pyramidal neuron labeling in the bilateral cortices were significantly correlated with behavioral recovery (P<0.05). CONCLUSIONS: Spontaneous functional recovery after stroke may, at least in part, be attributed to neuronal remodeling in the corticospinal system.


Subject(s)
Behavior, Animal/physiology , Pyramidal Tracts/pathology , Recovery of Function/physiology , Stroke/pathology , Stroke/physiopathology , Animals , Axons , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Fluorescent Antibody Technique , Herpesvirus 1, Suid , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Pyramidal Cells/growth & development , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Pyramidal Tracts/metabolism , Stroke/metabolism , Synapses/metabolism , Synapses/pathology
9.
J Neurochem ; 108(1): 231-45, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19094064

ABSTRACT

Doublecortin (DCX) is a microtubule (MT) binding protein that induces growth arrest at the G2-M phase of cell cycle in glioma and suppresses tumor xenograft in immunocompromised hosts. DCX expression was found in neuronal cells, but lacking in glioma cells. We tested the hypothesis that DCX inhibits glioma U87 cell mitosis and invasion. Our data showed that DCX synthesizing U87 cells underwent mitotic MT spindle catastrophe in a neurabin II dependent pathway. Synthesis of both DCX and neurabin II were required to induce apoptosis in U87 and human embryonic kidney 293T cells. In DCX expressing U87 cells, association of phosphorylated DCX with protein phosphatase-1 (PP1) in the cytosol disrupted the interaction between kinesin-13 and PP1 in the nucleus and yielded spontaneously active kinesin-13. The activated kinesin-13 caused mitotic MT catastrophe in spindle checkpoint. Phosphorylated-DCX induced depolymerization of actin filaments in U87 cells, down-regulated matrix metalloproteinases-2 and -9, and inhibited glioma U87 cell invasion in a neurabin II dependent pathway. Thus, localization of the DCX-neurabin II-PP1 complex in the cytosol of U87 tumor cells inhibited PP1 phosphatase activities leading to anti-glioma effects via (1) mitotic MT spindle catastrophe that blocks mitosis and (2) depolymerization of actin that inhibits glioma cell invasion.


Subject(s)
Gene Expression Regulation, Neoplastic , Glioma/metabolism , Microtubule-Associated Proteins/metabolism , Mitosis , Neuropeptides/metabolism , Spindle Apparatus/physiology , Actin Cytoskeleton , Adult , Analysis of Variance , Animals , Brain/metabolism , Cell Line , Cell Movement , Cell Proliferation , Cells, Cultured , Doublecortin Domain Proteins , Doublecortin Protein , Flow Cytometry , Humans , In Situ Nick-End Labeling/methods , Kinesins/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Nude , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/genetics , Mitosis/physiology , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Neuropeptides/genetics , Protein Phosphatase 1/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Transfection/methods , Tumor Cells, Cultured , Young Adult
10.
J Neurosci ; 27(12): 3157-62, 2007 Mar 21.
Article in English | MEDLINE | ID: mdl-17376977

ABSTRACT

Ischemic stroke induces neurogenesis in the subventricular zone (SVZ), and newly generated neurons in the SVZ migrate toward the ischemic boundary. However, the characteristics of migrating SVZ cells have not been investigated after stroke. Using time-lapse imaging in both SVZ cells and organotypic brain slice cultures, we measured the dynamics of SVZ cell division and migration of adult rats subjected to stroke. In normal brain slices, SVZ cells primarily migrated dorsally and ventrally along the lateral ventricular surface. However, in stroke brain slices, SVZ cells migrated laterally toward the striatal ischemic boundary. Cultured stroke-derived SVZ cells exhibited a significant (p < 0.01) increase in the migration distance (212 +/- 21 microm) compared with the nonstroke-derived SVZ cells (97 +/- 12 microm). Migrating stroke-derived SVZ cells spent significantly (p = 0.01) less time in cytokinesis (0.63 +/- 0.04 h) compared with the time (1.09 +/- 0.09 h) for nonstroke-derived SVZ cells. Newborn cells with a single leading process exhibited fast migration (7.2 +/- 0.8 microm/h), and cells with multiple processes showed stationary migration (3.6 +/- 0.8 microm/h). Stroke SVZ daughter cells further divided during their migration. The morphology of doublecortin (DCX)-positive cells in fixed brain sections resembled those observed in cultured newborn cells, and the DCX-positive cells proliferated in the ischemic striatum. Collectively, the present study suggests that stroke promotes cytokinesis of migrating neuroblasts, and these cells migrate toward the ischemic striatum with distinct migratory behaviors and retain the capacity for cell division during migration.


Subject(s)
Brain Ischemia/pathology , Cell Division/physiology , Cell Movement/physiology , Neurons/cytology , Neurons/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Cerebral Ventricles/cytology , Cerebral Ventricles/physiology , Corpus Striatum/blood supply , Corpus Striatum/cytology , Corpus Striatum/physiology , Doublecortin Protein , Male , Rats , Rats, Wistar , Stem Cells/cytology , Stem Cells/physiology
11.
Stroke ; 39(10): 2830-6, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18669895

ABSTRACT

BACKGROUND AND PURPOSE: Using a model of embolic stroke, the present study tested the hypothesis that blockage of endothelin-1 with S-0139, a specific endothelin type A receptor (ET(A)) antagonist, enhances the neuroprotective effect of recombinant tissue plasminogen activator (rtPA) by suppressing molecules that mediate thrombosis and blood brain barrier (BBB) disruption induced by ischemia and rtPA. METHODS: Rats (n=104) subjected to embolic middle cerebral artery (MCA) occlusion were randomly divided into 1 of 4 infusion groups with 26 rats per group: (1) the control group in which rats were administered saline, (2) the monotherapy rtPA group in which rtPA was intravenously administered at a dose of 10 mg/kg 4 hours after MCA occlusion, (3) the monotherapy S-0139 group in which S-0139 was intravenously given 2 hours after MCA occlusion, and (4) the combination of rtPA +S-0139 group in which S-0139 and rtPA were given 2 and 4 hours after MCA occlusion, respectively. Measurements of infarct volume and parenchymal hemorrhage, behavioral outcome, and immunostaining were performed on rats euthanized 1 and 7 days after stroke. RESULTS: The combination therapy of S-0139 and rtPA significantly (P<0.01) reduced infarct volume (24.8+/-0.9% versus 33.8+/-1.5% in control) and hemorrhagic area (7.1+/-6.1 microm(2) versus 36.5+/-19.2 microm(2) in control) and improved functional recovery compared with control saline-treated animals. Immunostaining analysis revealed that the combination therapy had the synergistically suppressed ischemia- and rtPA-induced ICAM-1, protease-activated receptor 1 (PAR-1), as well as accumulation of platelets in cerebral microvessels. Furthermore, the combination treatment synergistically reduced loss of laminin, ZO1, and occludin in cerebral vessels. CONCLUSIONS: These data suggest that S-0139 provides the neuroprotection by suppressing ischemia- and rtPA-triggered molecules that evoke thrombosis and BBB disruption.


Subject(s)
Caffeic Acids/administration & dosage , Fibrinolytic Agents/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/administration & dosage , Oleanolic Acid/analogs & derivatives , Receptor, PAR-1/drug effects , Tissue Plasminogen Activator/administration & dosage , Animals , Blood Platelets/drug effects , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/pathology , Collagen Type IV/biosynthesis , Collagen Type IV/drug effects , Drug Synergism , Endothelin Receptor Antagonists , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/drug effects , Male , Oleanolic Acid/administration & dosage , Rats , Rats, Wistar , Receptor, PAR-1/biosynthesis , Recovery of Function/drug effects , Tight Junctions/drug effects
12.
J Cereb Blood Flow Metab ; 28(4): 764-71, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17971789

ABSTRACT

Angiogenesis and neurogenesis are coupled processes. Using a coculture system, we tested the hypothesis that cerebral endothelial cells activated by ischemia enhance neural progenitor cell proliferation and differentiation, while neural progenitor cells isolated from the ischemic subventricular zone promote angiogenesis. Coculture of neural progenitor cells isolated from the subventricular zone of the adult normal rat with cerebral endothelial cells isolated from the stroke boundary substantially increased neural progenitor cell proliferation and neuronal differentiation and reduced astrocytic differentiation. Conditioned medium harvested from the stroke neural progenitor cells promoted capillary tube formation of normal cerebral endothelial cells. Blockage of vascular endothelial growth factor receptor 2 suppressed the effect of the endothelial cells activated by stroke on neurogenesis as well as the effect of the supernatant obtained from stroke neural progenitor cells on angiogenesis. These data suggest that angiogenesis couples to neurogenesis after stroke and vascular endothelial growth factor likely mediates this coupling.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Neurons/metabolism , Stem Cells/metabolism , Stroke/physiopathology , Animals , Blotting, Western , Caspase 3/metabolism , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , In Situ Nick-End Labeling , Male , Neurons/cytology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology
13.
Neuropharmacology ; 55(3): 345-52, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18632119

ABSTRACT

The subventricular zone (SVZ) of the lateral ventricle contains neural stem and progenitor cells that generate neuroblasts, which migrate to the olfactory bulb where they differentiate into interneurons. Ischemic stroke induces neurogenesis in the SVZ and these cells migrate to the boundary of the ischemic lesion. This article reviews current data on cytokinetics, signaling pathways and vascular niche that are involved in processes of proliferation, differentiation, and migration of neural progenitor cells after stroke.


Subject(s)
Brain Ischemia/pathology , Lateral Ventricles/pathology , Stroke/pathology , Animals , Brain Ischemia/complications , Cell Proliferation , Humans , Neuroglia/pathology , Neurons/pathology , Signal Transduction/physiology , Stem Cells/pathology , Stroke/etiology
14.
J Neurosci Methods ; 167(2): 376-83, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-17928064

ABSTRACT

Cell kinetics employed for embryonic models was modified and used to study the neuronogenesis in the subventricular zone (SVZ) in adult rats subjected to stroke. Enhanced analytical approaches were introduced and used to compare the cell cycle length (T(C)) and length in G(1) phase, T(G1), at various times after stroke to study the correlation between T(G1) and T(C) and to compare cell cycle evolution and proliferation profiles between the stroke and embryonic models. Our data indicate that cell cycle kinetics for the embryonic model can be applied to stroke in the adult. Significant reduction of T(G1) early after stroke (p<0.05) corresponds to an increase of neural progenitor cells remaining in the cycle at early times and cells exiting at later times. T(G1) correlates with T(C) (r=0.99, p<0.05). In conclusion, the analytical approaches proposed can be used to study the cell proliferation profiles in adult rats subjected to stroke with and without stroke therapy. The cell kinetics the cell proliferation profile differs between the stroke and embryonic models. T(C) evolution is three-fold slower in the cells and leave the cycle earlier and more frequently in the stroke model, compared to the embryonic model. T(C) is a surrogate measure of T(G1).


Subject(s)
Cell Cycle/physiology , Cell Proliferation , Infarction, Middle Cerebral Artery/pathology , Neurons/physiology , Stem Cells/physiology , Age Factors , Animals , Bromodeoxyuridine/metabolism , Cell Differentiation/physiology , Cell Division , Disease Models, Animal , Embryo, Mammalian , Lateral Ventricles/pathology , Male , Models, Neurological , Rats , Rats, Wistar
15.
Brain Res ; 1226: 18-26, 2008 Aug 21.
Article in English | MEDLINE | ID: mdl-18598677

ABSTRACT

The chemokine receptor CXCR4 and its ligand, stromal cell derived factor-1 alpha (SDF1 alpha) regulate neuroblast migration towards the ischemic boundary after stroke. Using loss- and gain-function, we investigated the biological effect of CXCR4/SDF1 alpha on neural progenitor cells. Neural progenitor cells, from the subventricular zone (SVZ) of the adult rat, were transfected with rat CXCR4-pLEGFP-C1 and pSIREN-RetroQ-CXCR4-siRNA retroviral vectors. Migration assay analysis showed that inhibition of CXCR4 by siRNA significantly reduced cell migration compared to the empty vector, indicating that CXCR4 mediated neural progenitor cell motility. When neural progenitor cells were cultured in growth medium containing bFGF (20 ng/ml), over-expression of CXCR4 significantly reduced the cell proliferation as measured by the number of bromodeoxyuridine+ (BrdU+) cells (26.4%) compared with the number in the control group (54.0%). Addition of a high concentration of SDF1 alpha (500 ng/ml) into the progenitor cells with over-expression of CXCR4 reversed the cell proliferation back to the control levels (57.6%). Immunostaining analysis showed that neither over-expression nor inhibition of CXCR4 altered the population of neurons and astrocytes, when neural progenitor cells were cultured in differentiation medium. These in vitro results suggest that CXCR4/SDF1 alpha primarily regulates adult neural progenitor cell motility but not differentiation, while over-expression of CXCR4 in the absence of SDF1 alpha decreases neural progenitor cell proliferation.


Subject(s)
Adult Stem Cells/drug effects , Cerebral Ventricles/cytology , Chemokine CXCL12/pharmacology , Neurons/drug effects , Receptors, CXCR4/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bromodeoxyuridine/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Fibroblast Growth Factors/pharmacology , Gene Expression/drug effects , Gene Expression/physiology , Gene Expression Profiling/methods , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Oligonucleotide Array Sequence Analysis/methods , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Receptors, CXCR4/genetics , Transfection/methods
16.
Curr Mol Med ; 7(5): 459-62, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17691960

ABSTRACT

Focal cerebral ischemia induces neurogenesis in the subventricular zone (SVZ) of the adult human brain. Neurogenesis is controlled by proliferation, differentiation, and migration of neural progenitor cells. This article reviews emerging data that changes of cell cycle kinetics of neural progenitor cells induced by stroke contribute to increased neural progenitor cell proliferation and that gene profiles control proliferation, differentiation, and migration of neural progenitor cells within the SVZ niche. A better understanding of gene profiles that control the biological function of adult SVZ neural progenitor cells could lead to more selective and effective treatments to enhance neurogenesis during stroke recovery.


Subject(s)
Brain Ischemia/pathology , Cell Differentiation , Cerebral Ventricles/cytology , Cerebral Ventricles/metabolism , Gene Expression Profiling , Neurons/cytology , Stem Cells/cytology , Adult , Animals , Cell Movement , Cell Proliferation , Humans
17.
J Neurosci ; 26(22): 5996-6003, 2006 May 31.
Article in English | MEDLINE | ID: mdl-16738242

ABSTRACT

We investigated the hypothesis that endothelial cells activated by erythropoietin (EPO) promote the migration of neuroblasts. This hypothesis is based on observations in vivo that treatment of focal cerebral ischemia with EPO enhances the migration of neuroblasts to the ischemic boundary, a site containing activated endothelial cells and angiogenic microvasculature. To model the microenvironment within the ischemic boundary zone, we used a coculture system of mouse brain endothelial cells (MBECs) and neural progenitor cells derived from the subventricular zone of the adult mouse. Treatment of MBECs with recombinant human EPO (rhEPO) significantly increased secretion of matrix metalloproteinase 2 (MMP2) and MMP9. rhEPO-treated MBEC supernatant as conditioned medium significantly increased the migration of neural progenitor cells. Application of an MMP inhibitor abolished the supernatant-enhanced migration. Incubation of neurospheres alone with rhEPO failed to increase progenitor cell migration. rhEPO activated phosphatidylinositol 3-kinase/Akt (PI3K/Akt) and extracellular signal-regulated kinase (ERK1/2) in MBECs. Selective inhibition of the PI3K/Akt and ERK1/2 pathways significantly attenuated the rhEPO-induced MMP2 and MMP9, which suppressed neural progenitor cell migration promoted by the rhEPO-activated MBECs. Collectively, our data show that rhEPO-activated endothelial cells enhance neural progenitor cell migration by secreting MMP2 and MMP9 via the PI3K/Akt and ERK1/2 signaling pathways. These data demonstrate that activated endothelial cells can promote neural progenitor cell migration, and provide insight into the molecular mechanisms underlying the attraction of newly generated neurons to injured areas in brain.


Subject(s)
Endothelium, Vascular/physiology , Erythropoietin/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neurons/physiology , Stem Cells/physiology , Animals , Cell Movement/drug effects , Cell Movement/physiology , Cerebrovascular Circulation , Coculture Techniques , Endothelium, Vascular/drug effects , Genes, Reporter , Humans , Male , Mice , Neurons/drug effects , Rats , Rats, Wistar , Recombinant Proteins , Stem Cells/drug effects
18.
J Cereb Blood Flow Metab ; 27(3): 564-74, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16835628

ABSTRACT

Neural progenitor cells in the subventricular zone (SVZ) of the lateral ventricular wall give rise to new neurons throughout rodent life. Ischemic stroke induces angiogenesis and neurogenesis. Using laser capture microdissection (LCM) in combination with microarrays containing approximately 400 known genes associated with stem cells and angiogenesis, we investigated gene profiles of SVZ cells in the adult mouse subjected to middle cerebral artery occlusion. Our data revealed that nonstroke SVZ cells expressed sets of genes that are important for neural progenitor cell proliferation, differentiation, and migration. In addition, stroke SVZ cells expressed many genes involved in neurogenesis during embryonic development but were not detected in nonstroke SVZ cells. Stroke upregulated genes were verified by real-time reverse transcriptase-polymerase chain reaction and immunostaining. These data indicate that adult SVZ cells recapture embryonic molecular signals after stroke and provide insight into the molecular mechanisms, which regulate the biological function of neural progenitor cells in the SVZ of adult rodent brain under physiological and stroke conditions.


Subject(s)
Gene Expression , Lateral Ventricles/cytology , Neovascularization, Pathologic/genetics , Neurons/physiology , Stem Cells/physiology , Stroke/physiopathology , Animals , Gene Expression Profiling , Immunohistochemistry , In Situ Hybridization , Lasers , Male , Mice , Microdissection , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
19.
J Cereb Blood Flow Metab ; 27(6): 1201-12, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17200679

ABSTRACT

Adult ependymal cells are postmitotic and highly differentiated. Radial glial cells are neurogenic precursors. Here, we show that stroke acutely stimulated adult ependymal cell proliferation, and dividing ependymal cells of the lateral ventricle had genotype, phenotype, and morphology of radial glial cells in the rat. The majority of radial glial cells exhibited symmetrical division about the cell cleavage plane, and a radial fiber was maintained throughout each stage of cell mitosis. Increases of radial glial cells parallel expansion of neural progenitors in the subventricular zone (SVZ). Furthermore, after stroke radial glial cells derived from the SVZ supported neuron migration. These results indicate that adult ependymal cells divide and transform into radial glial cells after stroke, which could function as neural progenitor cells to generate new neurons and act as scaffolds to support neuroblast migration towards the ischemic boundary region.


Subject(s)
Ependyma/physiology , Lateral Ventricles/pathology , Neuroglia/physiology , Stroke/pathology , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Ependyma/cytology , Mitosis , Neuroglia/cytology , Neurons/cytology , Rats , Stem Cells/cytology , Stem Cells/physiology
20.
J Neurosci Methods ; 162(1-2): 91-100, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17287025

ABSTRACT

BACKGROUND AND PURPOSE: Ischemic stroke induces spreading depression of brain waves and ischemic depolarizations, suggesting electrical activity of neurons is sensitive to stroke. The present study was designed to measure the electrophysiological response of an array of individual neurons to ischemic stroke in rats. METHODS: A custom-made microwire electrode array (16 channels) was implanted in the cortical area supplied by the middle cerebral artery, spanning the core and boundary of the ischemic lesion. The electrophysiological activity of individual neurons was simultaneously recorded before, during and one week after middle cerebral artery occlusion (MCAo). RESULTS: Neuronal activities were significantly reduced immediately after MCAo. Intermittent silent periods (SP) appeared within minutes or hours after MCAo and lasted variable times. Between intermittent SP, neurons fired irregular bursting spikes (BS) with small magnitudes. Intermittent SP and irregular BS progressed in one day post stroke to persistent SP in channels close to the ischemic core or to regular BS with small amplitudes in the penumbral zone. Both persistent SP and regular BS persisted for at least seven days. CONCLUSIONS: Electrode array can be used to simultaneously record multiple individual neurons in response to ischemic stroke. This study provides the first evidence that the primary electrophysiological activity of multiple individual neurons to ischemic stroke is reduced in the lesion boundary and/or stopped in and adjacent to the lesion core.


Subject(s)
Cerebral Cortex/physiology , Electrophysiology/methods , Middle Cerebral Artery/physiology , Neurons/physiology , Animals , Arterial Occlusive Diseases/physiopathology , Cerebral Cortex/physiopathology , Electrophysiology/instrumentation , Male , Microelectrodes , Middle Cerebral Artery/physiopathology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL