Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Exp Neurol ; 297: 14-24, 2017 11.
Article in English | MEDLINE | ID: mdl-28716558

ABSTRACT

Stimulation of endogenous neurogenesis and recruitment of neural progenitors from the subventricular zone (SVZ) neurogenic site may represent a useful strategy to improve regeneration in the ischemic cortex. Here, we tested whether transgenic overexpression of extracellular matrix metalloproteinase inducer (EMMPRIN), the regulator of matrix metalloproteinases (MMPs) expression, in endogenous neural progenitor cells (NPCs) in the subventricular zone (SVZ) could increase migration towards ischemic injury. For this purpose, we applied a lentivector-mediated gene transfer system. We found that EMMPRIN-transduced progenitors exhibited enhanced MMP-2 activity in vitro and showed improved motility in 3D collagen gel as well as in cortical slices. Using a rat model of neonatal ischemia, we showed that EMMPRIN overexpressing SVZ cells invade the injured cortical tissue more efficiently than controls. Our results suggest that EMMPRIN overexpression could be suitable approach to improve capacities of endogenous or transplanted progenitors to invade the injured cortex.


Subject(s)
Basigin/biosynthesis , Brain Ischemia/metabolism , Cell Movement/physiology , Cerebral Cortex/metabolism , Lateral Ventricles/metabolism , Neural Stem Cells/metabolism , Animals , Animals, Newborn , Basigin/genetics , Brain Ischemia/pathology , Cerebral Cortex/pathology , Gene Expression , Lateral Ventricles/pathology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Rats, Wistar
2.
Mol Neurodegener ; 12(1): 15, 2017 02 13.
Article in English | MEDLINE | ID: mdl-28193235

ABSTRACT

BACKGROUND: Alzheimer's disease (AD)-linked protein, presenilin 1 (PS1), is present at the synapse, and the knock-out of presenilin in mice leads to synaptic dysfunction. On the other hand, synaptic activity was shown to influence PS1-dependent generation of distinct amyloid ß (Aß) species. However, the precise nature of these regulations remains unclear. The current study reveals novel role of PS1 at the synapse, and deciphers how PS1 and synaptic vesicle-associated protein, synaptotagmin 1 (Syt1) modulate each other functions in neurons via direct activity-triggered interaction. Additionally, the therapeutic potential of fostering PS1-Syt1 binding is investigated as a synapse-specific strategy for AD prevention. METHODS: PS1-based cell-permeable peptide targeting PS1-Syt1 binding site was designed to inhibit PS1-Syt1 interaction in neurons. PS1 conformation, synaptic vesicle exocytosis and trafficking were assayed by fluorescence lifetime imaging microscopy (FLIM), glutamate release/synaptopHluorin assay, and fluorescence recovery after photobleaching, respectively. Syt1 level and interaction with PS1 in control and sporadic AD brains were determined by immunohistochemistry and FLIM. AAV-mediated delivery of Syt1 into mouse hippocampi was used to investigate the therapeutic potential of strengthening PS1-Syt1 binding in vivo. Statistical significance was determined using two-tailed unpaired Student's t-test, Mann-Whitney's U-test or two-way ANOVA followed by a Bonferroni's post-test. RESULTS: We demonstrate that targeted inhibition of the PS1-Syt1 binding in neurons, without changing the proteins' expression level, triggers "pathogenic" conformational shift of PS1, and consequent increase in the Aß42/40 ratio. Moreover, our data indicate that PS1, by binding directly to Syt1, regulates synaptic vesicle trafficking and facilitates exocytosis and neurotransmitter release. Analysis of human brain tissue revealed that not only Syt1 levels but also interactions between remaining Syt1 and PS1 are diminished in sporadic AD. On the other hand, overexpression of Syt1 in mouse hippocampi was found to potentiate PS1-Syt1 binding and promote "protective" PS1 conformation. CONCLUSIONS: The study reports novel functions of PS1 and Syt1 at the synapse, and demonstrates the importance of PS1-Syt1 binding for exocytosis and safeguarding PS1 conformation. It suggests that reduction in the Syt1 level and PS1-Syt1 interactions in AD brain may present molecular underpinning of the pathogenic PS1 conformation, increased Aß42/40 ratio, and impaired exocytosis.


Subject(s)
Amyloid beta-Peptides/metabolism , Exocytosis/physiology , Presenilin-1/metabolism , Synaptotagmin I/metabolism , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cells, Cultured , Humans , Immunohistochemistry/methods , Mice, Inbred C57BL , Neurons/metabolism , Synapses/metabolism
3.
Brain ; 140(3): 692-706, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28115364

ABSTRACT

Transplanted neurons derived from stem cells have been proposed to improve function in animal models of human disease by various mechanisms such as neuronal replacement. However, whether the grafted neurons receive functional synaptic inputs from the recipient's brain and integrate into host neural circuitry is unknown. Here we studied the synaptic inputs from the host brain to grafted cortical neurons derived from human induced pluripotent stem cells after transplantation into stroke-injured rat cerebral cortex. Using the rabies virus-based trans-synaptic tracing method and immunoelectron microscopy, we demonstrate that the grafted neurons receive direct synaptic inputs from neurons in different host brain areas located in a pattern similar to that of neurons projecting to the corresponding endogenous cortical neurons in the intact brain. Electrophysiological in vivo recordings from the cortical implants show that physiological sensory stimuli, i.e. cutaneous stimulation of nose and paw, can activate or inhibit spontaneous activity in grafted neurons, indicating that at least some of the afferent inputs are functional. In agreement, we find using patch-clamp recordings that a portion of grafted neurons respond to photostimulation of virally transfected, channelrhodopsin-2-expressing thalamo-cortical axons in acute brain slices. The present study demonstrates, for the first time, that the host brain regulates the activity of grafted neurons, providing strong evidence that transplanted human induced pluripotent stem cell-derived cortical neurons can become incorporated into injured cortical circuitry. Our findings support the idea that these neurons could contribute to functional recovery in stroke and other conditions causing neuronal loss in cerebral cortex.


Subject(s)
Brain Injuries/surgery , Evoked Potentials, Somatosensory/physiology , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Synapses/physiology , Action Potentials , Afferent Pathways/physiology , Animals , Brain/cytology , Brain/ultrastructure , Brain Injuries/etiology , Cell Line, Transformed , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/ultrastructure , Disease Models, Animal , Humans , Lysine/analogs & derivatives , Lysine/metabolism , Male , Neurons/physiology , Neurons/ultrastructure , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Nude , Rats, Sprague-Dawley , Stroke/complications , Synapses/ultrastructure , Ventral Thalamic Nuclei/cytology
4.
Microsc Res Tech ; 79(6): 557-64, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27087359

ABSTRACT

We investigated the ultrastructural characteristics of mouse adipose-derived stem/stromal cells (ASCs) induced towards osteogenic lineage. ASCs were isolated from adipose tissue of FVB-Cg-Tg(GFPU)5Nagy/J mice and expanded in monolayer culture. Flow cytometry, histochemical staining, and electron microscopy techniques were used to characterize the ASCs with respect to their ability for osteogenic differentiation capacity. Immunophenotypically, ASCs were characterized by high expression of the CD44 and CD90 markers, while the relative content of cells expressing CD45, CD34 and CD117 markers was <2%. In assays of differentiation, the positive response to osteogenic differentiation factors was observed and characterized by deposition of calcium in the extracellular matrix and alkaline phosphatase production. Electron microscopy analysis revealed that undifferentiated ASCs had a rough endoplasmic reticulum with dilated cisterns and elongated mitochondria. At the end of the osteogenic differentiation, the ASCs transformed from their original fibroblast-like appearance to having a polygonal osteoblast-like morphology. Ultrastructurally, these cells were characterized by large euchromatic nucleus and numerous cytoplasm containing elongated mitochondria, a very prominent rough endoplasmic reticulum, Golgi apparatus and intermediate filament bundles. Extracellular matrix vesicles of variable size similar to the calcification nodules were observed among collagen fibrils. Our data provide the ultrastructural basis for further studies on the cellular mechanisms involved in osteogenic differentiation of mouse adipose-derived stem/stromal cells. Microsc. Res. Tech. 79:557-564, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/physiology , Osteogenesis/physiology , Stromal Cells/ultrastructure , Animals , Antigens, CD , Cells, Cultured , Flow Cytometry , Immunophenotyping , Mice , Microscopy, Electron, Transmission , Stromal Cells/chemistry , Stromal Cells/cytology
5.
Cell Calcium ; 59(5): 251-61, 2016 05.
Article in English | MEDLINE | ID: mdl-26969192

ABSTRACT

Pyramidal neurons of the hippocampus possess differential susceptibility to the ischemia-induced damage with the highest vulnerability of CA1 and the lower sensitivity of CA3 neurons. This damage is triggered by Ca(2+)-dependent excitotoxicity and can result in a delayed cell death that might be potentially suspended through activation of endogenous neuroprotection with the hypoxia-inducible transcription factors (HIF). However, the molecular mechanisms of this neuroprotection remain poorly understood. Here we show that prolonged (30min) oxygen and glucose deprivation (OGD) in situ impairs intracellular Ca(2+) regulation in CA1 rather than in CA3 neurons with the differently altered expression of genes coding Ca(2+)-ATPases: the mRNA level of plasmalemmal Ca(2+)-ATPases (PMCA1 and PMCA2 subtypes) was downregulated in CA1 neurons, whereas the mRNA level of the endoplasmic reticulum Ca(2+)-ATPases (SERCA2b subtype) was increased in CA3 neurons at 4h of re-oxygenation after prolonged OGD. These demonstrate distinct susceptibility of CA1 and CA3 neurons to the ischemic impairments in intracellular Ca(2+) regulation and Ca(2+)-ATPase expression. Stabilization of HIF-1α by inhibiting HIF-1α hydroxylation prevented the ischemic decrease in both PMCA1 and PMCA2 mRNAs in CA1 neurons, upregulated the SERCA2b mRNA level and eliminated the OGD-induced Ca(2+) store dysfunction in these neurons. Cumulatively, these findings reveal the previously unknown HIF-1α-driven upregulation of Ca(2+)-ATPases as a mechanism opposing the ischemic impairments in intracellular Ca(2+) regulation in hippocampal neurons. The ability of HIF-1α to modulate expression of genes coding Ca(2+)-ATPases suggests SERCA2b as a novel target for HIF-1 and may provide potential implications for HIF-1α-stabilizing strategy in activating endogenous neuroprotection.


Subject(s)
Calcium/metabolism , Hippocampus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neurons/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Cell Death/drug effects , Cytoplasm/metabolism , Down-Regulation/drug effects , Ischemia/metabolism , Neuroprotective Agents/pharmacology , Rats, Wistar , Transcriptional Activation/drug effects , Up-Regulation/drug effects
6.
Cell Transplant ; 25(7): 1359-69, 2016.
Article in English | MEDLINE | ID: mdl-26810970

ABSTRACT

Neural progenitor cells (NPCs) overexpressing fibroblast growth factor 2 (FGF-2) have the distinct tendency to associate with the vasculature and establish multiple proliferative clusters in the perivascular environment after transplantation into the cerebral cortex. Strikingly, the vascular clusters of progenitor cells give rise to immature neurons after ischemic injury, raising prospects for the formation of ectopic neurogenic niches for repair. We investigated the spatial relationship of perivascular clusters with the host vascular structures. FGF-2-GFP-transduced NPCs were transplanted into the intact somatosensory rat cortex. Confocal microscopic analysis revealed that grafted cells preferentially contacted venules at sites with aquaporin-4-positive astrocytic endfeet and avoided contacts with desmin-positive pericytes. Electron microscopic analysis confirmed that grafted cells preferentially made contact with astroglial endfeet, and only a minority of them reached the endothelial basal lamina. These results provide new insights into the fine structural and anatomical relationship between grafted FGF-2-transduced NPCs and the host vasculature.


Subject(s)
Cerebral Cortex/blood supply , Fibroblast Growth Factor 2/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Animals , Animals, Newborn , Astrocytes/cytology , Blood Vessels/cytology , Blood Vessels/ultrastructure , Cell Aggregation , Cells, Cultured , Cerebral Cortex/cytology , Female , Green Fluorescent Proteins/metabolism , Male , Pericytes/cytology , Rats, Sprague-Dawley , Somatosensory Cortex/cytology
7.
J Neurosci Res ; 92(8): 964-74, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24753232

ABSTRACT

The adult CNS has a very limited capacity to regenerate neurons after insult. To overcome this limitation, the transplantation of neural progenitor cells (NPCs) has developed into a key strategy for neuronal replacement. This study assesses the long-term survival, migration, differentiation, and functional outcome of NPCs transplanted into the ischemic murine brain. Hippocampal neural progenitors were isolated from FVB-Cg-Tg(GFPU)5Nagy/J transgenic mice expressing green fluorescent protein (GFP). Syngeneic GFP-positive NPCs were stereotactically transplanted into the hippocampus of FVB mice following a transient global cerebral ischemia model. Behavioral tests revealed that ischemia/reperfusion induced spatial learning disturbances in the experimental animals. The NPC transplantation promoted cognitive function recovery after ischemic injury. To study the long-term fate of grafted GFP-positive NPCs in a host brain, immunohistochemical approaches were applied. Confocal microscopy revealed that grafted cells survived in the recipient tissue for 90 days following transplantation and differentiated into mature neurons with extensive dendritic trees and apparent spines. Immunoelectron microscopy confirmed the formation of synapses between the transplanted GFP-positive cells and host neurons that may be one of the factors underlying cognitive function recovery. Repair and functional recovery following brain damage represent a major challenge for current clinical and basic research. Our results provide insight into the therapeutic potential of transplanted hippocampal progenitor cells following ischemic brain injury.


Subject(s)
Brain Ischemia/therapy , Hippocampus/pathology , Nerve Degeneration/pathology , Neural Stem Cells/transplantation , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Hippocampus/physiopathology , Maze Learning/physiology , Mice , Nerve Degeneration/physiopathology , Neural Stem Cells/pathology , Stem Cell Transplantation , Synapses/pathology
8.
Int J Dev Neurosci ; 35: 64-71, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24642047

ABSTRACT

The first milk, colostrum, is an important source of nutrients and an exclusive source of immunoglobulins (Ig), essential for the growth and protection from infection of newborn pigs. Colostrum intake has also been shown to affect the vitality and behaviour of neonatal pigs. The objective of this study was to evaluate the effects of feeding colostrum and plasma immunoglobulin on brain development in neonatal pigs. Positive correlations were found between growth, levels of total protein and IgG in blood plasma and hippocampus development in sow-reared piglets during the first 3 postnatal days. In piglets fed an elemental diet (ED) for 24h, a reduced body weight, a lower plasma protein level and a decreased level of astrocyte specific protein in the hippocampus was observed, as compared to those that were sow-reared. The latter was coincident with a reduced microgliogenesis and an essentially diminished number of neurons in the CA1 area of the hippocampus after 72h. Supplementation of the ED with purified plasma Ig, improved the gliogenesis and supported the trophic and immune status of the hippocampus. The data obtained indicate that the development of the hippocampus structure is improved by colostrum or an Ig-supplemented elemental diet in order to stimulate brain protein synthesis and its development during the early postnatal period.


Subject(s)
Colostrum , Hippocampus/growth & development , Immunoglobulin G/administration & dosage , Immunoglobulin G/blood , Swine/blood , Swine/growth & development , Administration, Oral , Animals , Animals, Newborn , Body Weight/drug effects , Body Weight/physiology , Dietary Supplements , Female , Hippocampus/cytology , Hippocampus/drug effects , Male , Nerve Tissue Proteins/metabolism , Organ Size/drug effects , Organ Size/physiology
9.
Pharmacol Rep ; 63(3): 840-4, 2011.
Article in English | MEDLINE | ID: mdl-21857096

ABSTRACT

Negatively charged sialic acid residues located close to pores of voltage-gated sodium channels substantially influence their gating properties. The in vitro low Mg²âº seizure model is used to emulate difficult-to-treat status epilepticus. Using this model on cultured hippocampal slices, we examined the effectiveness of desialylation in reducing persistent seizure-like activity. We show that desialylation in cultured hippocampal slices effectively suppresses seizure-like activity induced by low Mg²âº. These findings suggest that targeting negatively charged sialic acids may be an effective strategy to treat status epilepticus.


Subject(s)
Hippocampus/pathology , Neuraminidase/pharmacology , Sialic Acids/metabolism , Status Epilepticus/drug therapy , Animals , Disease Models, Animal , Drug Delivery Systems , Magnesium/metabolism , Rats , Rats, Wistar , Status Epilepticus/physiopathology
10.
Brain Res ; 1386: 175-83, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21338581

ABSTRACT

It is well known that a brief anoxia or hypoxia episodes can render brain resistant to a subsequent ischemia. Recent investigations indicate that mechanisms of such stimulated endogenous neuroprotection are related to the family of hypoxia-inducible factors (HIF), however there are still little data available on the role of HIF family members in hippocampus-a brain structure, highly sensitive to oxygen deficiency. We have used the model of cultured hippocampal slices and single-cell quantitative RT-PCR to study HIF-1α and HIF-3α mRNA expression following triple 5-min mild anoxia, 30-min oxygen-glucose deprivation and their combination. We also tested the effects of HIF prolyl-hydroxylase inhibition with 2,4-pyridinedicarboxylic acid diethyl ester pre-treatment followed by a 30-min oxygen-glucose deprivation. It was found that neuronal damage induced by oxygen-glucose deprivation was accompanied by a significant decrease in both HIF-1α and HIF-3α mRNA levels in CA1 but not CA3 neurons. Anoxia preconditioning did not affect cell viability and HIF mRNA levels but applied before oxygen-glucose deprivation prevented neuronal damage and suppression of HIF-1α and HIF-3α mRNA expression. It was also found that effects of the prolyl-hydroxylase inhibitor were similar to anoxia preconditioning. These results suggest that anoxia preconditioning increases anti-ischemic neuronal resistance which to a certain extent correlates with the changes of HIF-1α and HIF-3α expression.


Subject(s)
CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Ischemia, Brain/therapy , Ischemic Preconditioning/methods , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Animals, Newborn , Brain Infarction/enzymology , Brain Infarction/physiopathology , Brain Infarction/therapy , CA1 Region, Hippocampal/drug effects , Disease Models, Animal , Hypoxia/enzymology , Hypoxia/physiopathology , Hypoxia/therapy , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Ischemia, Brain/enzymology , Hypoxia-Ischemia, Brain/physiopathology , Organ Culture Techniques , Procollagen-Proline Dioxygenase/metabolism , Rats , Rats, Wistar , Transcription Factors/genetics , Up-Regulation/physiology
11.
Neuropharmacology ; 60(5): 757-64, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21187106

ABSTRACT

Synaptic activity, such as long-term potentiation (LTP), has been shown to induce morphological plasticity of excitatory synapses on dendritic spines through the spine head and postsynaptic density (PSD) enlargement and reorganization. Much less, however, is known about activity-induced morphological modifications of inhibitory synapses. Using an in vitro model of rat organotypic hippocampal slice cultures and electron microscopy, we studied activity-related morphological changes of somatic inhibitory inputs triggered by a brief oxygen-glucose deprivation (OGD) episode, a condition associated with a synaptic enhancement referred to as anoxic LTP and a structural remodeling of excitatory synapses. Three-dimensional reconstruction of inhibitory axo-somatic synapses at different times before and after brief OGD revealed important morphological changes. The PSD area significantly and markedly increased at synapses with large and complex PSDs, but not at synapses with simple, macular PSDs. Activity-related changes of PSD size and presynaptic bouton volume developed in a strongly correlated manner. Analyses of single and serial sections further showed that the density of inhibitory synaptic contacts on the cell soma did not change within 1 h after OGD. In contrast, the proportion of the cell surface covered with inhibitory PSDs, as well as the complexity of these PSDs significantly increased, with less macular PSDs and more complex, segmented shapes. Together, these data reveal a rapid activity-related restructuring of somatic inhibitory synapses characterized by an enlargement and increased complexity of inhibitory PSDs, providing a new mechanism for a quick adjustment of the excitatory-inhibitory balance. This article is part of a Special Issue entitled 'Synaptic Plasticity & Interneurons'.


Subject(s)
CA1 Region, Hippocampal/physiology , Excitatory Postsynaptic Potentials/physiology , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , Synapses/pathology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/physiology , Animals , Cell Culture Techniques , Dendritic Spines/physiology , Hippocampus , Long-Term Potentiation/physiology , Post-Synaptic Density/physiology , Presynaptic Terminals/physiology , Rats , Synapses/physiology
12.
Eur J Neurosci ; 32(11): 1889-96, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21044183

ABSTRACT

Polysialic acids are widely distributed in neuronal tissue. Due to their position on glycoproteins and gangliosides on the outer cell membranes and anionic nature, polysialic acids are involved in multiple cell signaling events. The level of sialylation of the cellular surface is regulated by endogenous neuraminidase (NEU), which catalyses the hydrolysis of terminal sialic acid residues. Using the specific blocker of endogenous NEU, N-acetyl-2,3-dehydro-2-deoxyneuraminic acid (NADNA), we show that downregulation of the endogenous NEU activity causes a significant increase in the level of hippocampal tissue sialylation. Acute application of NADNA increased the firing frequency and amplitude of spontaneous synchronous oscillations, and frequency of multiple unit activity in cultured hippocampal slices. The tonic phase of seizure-like activity in the low-magnesium model of ictogenesis was significantly increased in slices pretreated with NADNA. These data indicate that the degree of synchronization is influenced by the amount of active NEU in cultured hippocampal slices. Pretreatment with NADNA led to an increase of the density of simple and perforated synapses in the hippocampal CA1 stratum radiatum region. Co-incubation of slices with NADNA and high concentrations of calcium eliminated the effect of the NEU blocker on synaptic density, suggesting that synaptogenesis observed following downregulation of the endogenous NEU activity is an activity-dependent process.


Subject(s)
Hippocampus/cytology , Hippocampus/physiology , Neuraminic Acids/metabolism , Neuraminidase/antagonists & inhibitors , Neurons/enzymology , Neurons/physiology , Synapses/physiology , Animals , Calcium/metabolism , Electrophysiology , N-Acetylneuraminic Acid/analogs & derivatives , N-Acetylneuraminic Acid/metabolism , Neuraminic Acids/chemistry , Neuraminidase/metabolism , Neurons/cytology , Rats , Rats, Wistar , Tissue Culture Techniques
13.
Stem Cells ; 27(6): 1309-17, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19489096

ABSTRACT

Stem/progenitor cell-based therapies hold promises for repairing the damaged nervous system. However, the efficiency of these approaches for neuronal replacement remains very limited. A major challenge is to develop pretransplant cell manipulations that may promote the survival, engraftment, and differentiation of transplanted cells. Here, we investigated whether overexpression of fibroblast growth factor-2 (FGF-2) in grafted neural progenitors could improve their integration in the host tissue. We show that FGF-2-transduced progenitors grafted in the early postnatal rat cortex have the distinct tendency to associate with the vasculature and establish multiple proliferative clusters in the perivascular environment. The contact with vessels appears to be critical for maintaining progenitor cells in an undifferentiated and proliferative phenotype in the intact cortex. Strikingly, perivascular clusters of FGF-2 expressing cells seem to supply immature neurons in an ischemic environment. Our data provide evidence that engineering neural progenitors to overexpress FGF-2 may be a suitable strategy to improve the integration of grafted neural progenitor cells with the host vasculature thereby generating neurovascular clusters with a neurogenic potential for brain repair.


Subject(s)
Fibroblast Growth Factor 2/biosynthesis , Hypoxia-Ischemia, Brain/surgery , Neurons/metabolism , Stem Cell Transplantation/methods , Stem Cells/metabolism , Animals , Blood Vessels , Cell Differentiation/physiology , Fibroblast Growth Factor 2/genetics , Immunohistochemistry , Neurons/cytology , Rats , Rats, Wistar , Stem Cells/cytology
14.
Hippocampus ; 19(8): 753-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19156853

ABSTRACT

Patterns of activity that induce synaptic plasticity at excitatory synapses, such as long-term potentiation, result in structural remodeling of the postsynaptic spine, comprising an enlargement of the spine head and reorganization of the postsynaptic density (PSD). Furthermore, spine synapses represent complex functional units in which interaction between the presynaptic varicosity and the postsynaptic spine is also modulated by surrounding astroglial processes. To investigate how activity patterns could affect the morphological interplay between these three partners, we used an electron microscopic (EM) approach and 3D reconstructions of excitatory synapses to study the activity-related morphological changes underlying induction of synaptic potentiation by theta burst stimulation or brief oxygen/glucose deprivation episodes in hippocampal organotypic slice cultures. EM analyses demonstrated that the typical glia-synapse organization described in in vivo rat hippocampus is perfectly preserved and comparable in organotypic slice cultures. Three-dimensional reconstructions of synapses, classified as simple or complex depending upon PSD organization, showed significant changes following induction of synaptic potentiation using both protocols. The spine head volume and the area of the PSD significantly enlarged 30 min and 1 h after stimulation, particularly in large synapses with complex PSD, an effect that was associated with a concomitant enlargement of presynaptic terminals. Furthermore, synaptic activity induced a pronounced increase of the glial coverage of both pre- and postsynaptic structures, these changes being prevented by application of the NMDA receptor antagonist D-2-amino-5-phosphonopentanoic acid. These data reveal dynamic, activity-dependent interactions between glial processes and pre- and postsynaptic partners and suggest that glia can participate in activity-induced structural synapse remodeling.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Neuroglia/physiology , Neurons/physiology , Synapses/physiology , Synaptic Transmission/physiology , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Cell Hypoxia/physiology , Dendritic Spines/drug effects , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Glucose/deficiency , Hippocampus/drug effects , Hippocampus/ultrastructure , Imaging, Three-Dimensional , In Vitro Techniques , Linear Models , Microelectrodes , Microscopy, Electron, Transmission , Neuroglia/drug effects , Neuroglia/ultrastructure , Neurons/drug effects , Neurons/ultrastructure , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synapses/drug effects , Synapses/ultrastructure
15.
Neurosci Lett ; 444(2): 181-5, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18708122

ABSTRACT

Diabetic patients show impaired brain functions, although underlying mechanisms remain unclear. Little is known as well about early diabetes-related changes in a brain tissue. To investigate them we analyzed the reaction of astroglial cells in the hippocampus of rats rendered diabetic by a single injection of streptozotocin (STZ). Astrocyte count, size and shape as well as levels of glial fibrillary acidic protein (GFAP) and S100b protein were assessed 3, 7 and 14 days after the STZ injection using immunohistochemistry, immuno-enzyme assay and computer-assisted image analysis. The reduced GFAP-positive cell count was found on day 3 when these cells were significantly smaller and less arborized with respect to the control. This tendency reversed on day 7 when more numerous GFAP-positive cells grew in size and became more ramified. S100b-positive cell count changes followed a contrasting pattern, elevating on days 3 and 7 and dropping on day 14. The level of cytoskeletal GFAP changed in parallel with GFAP expression revealed by immunocytochemistry, while cytosolic GFAP level started to increase only 7 days after the STZ injection. At the same time S100b level experienced an elevation on day 3 reaching the peak on day 7 and decreasing afterwards. These results suggest that the reaction of astroglial cells may be the earliest response of the brain tissue to an altered glucose metabolism playing presumably the critical role in the mechanisms underlying diabetes-related impairments of brain functions.


Subject(s)
Astrocytes/pathology , Diabetes Mellitus, Experimental/pathology , Hippocampus/pathology , Animals , Astrocytes/metabolism , Cell Count , Cell Shape , Cell Size , Diabetes Mellitus, Experimental/chemically induced , Glial Fibrillary Acidic Protein/metabolism , Immunoassay , Immunohistochemistry , Male , Nerve Growth Factors/metabolism , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Streptozocin
16.
Cell Calcium ; 43(6): 591-601, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18054077

ABSTRACT

Ca(2+) signaling is the astrocyte form of excitability and the endoplasmic reticulum (ER) plays an important role as an intracellular Ca(2+) store. Since the subcellular distribution of the ER influences Ca(2+) signaling, we compared the arrangement of ER in astrocytes of hippocampus tissue and astrocytes in cell culture by electron microscopy. While the ER was usually located in close apposition to the plasma membrane in astrocytes in situ, the ER in cultured astrocytes was close to the nuclear membrane. Activation of metabotropic receptors linked to release of Ca(2+) from ER stores triggered distinct responses in cultured and in situ astrocytes. In culture, Ca(2+) signals were commonly first recorded close to the nucleus and with a delay at peripheral regions of the cells. Store-operated Ca(2+) entry (SOC) as a route to refill the Ca(2+) stores could be easily identified in cultured astrocytes as the Zn(2+)-sensitive component of the Ca(2+) signal. In contrast, such a Zn(2+)-sensitive component was not recorded in astrocytes from hippocampal slices despite of evidence for SOC. Our data indicate that both, astrocytes in situ and in vitro express SOC necessary to refill stores, but that a SOC-related signal is not recorded in the cytoplasm of astrocytes in situ since the stores are close to the plasma membrane and the refill does not affect cytoplasmic Ca(2+) levels.


Subject(s)
Astrocytes/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Aniline Compounds , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/ultrastructure , Calcium/pharmacology , Calcium Signaling/drug effects , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cytoplasm/drug effects , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Fluorescent Dyes , Green Fluorescent Proteins , Hippocampus/metabolism , Hippocampus/ultrastructure , Immunohistochemistry , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Nuclear Envelope/drug effects , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Organ Culture Techniques , Staining and Labeling , Xanthenes , Zinc/metabolism , Zinc/pharmacology
17.
Hippocampus ; 16(10): 814-25, 2006.
Article in English | MEDLINE | ID: mdl-16892187

ABSTRACT

Relatively mild ischemic episode can initiate a chain of events resulting in delayed cell death and significant lesions in the affected brain regions. We studied early synaptic modifications after brief ischemia modeled in rats by transient vessels' occlusion in vivo or oxygen-glucose deprivation in vitro and resulting in delayed death of hippocampal CA1 pyramidal cells. Electron microscopic analysis of excitatory spine synapses in CA1 stratum radiatum revealed a rapid increase of the postsynaptic density (PSD) thickness and length, as well as formation of concave synapses with perforated PSD during the first 24 h after ischemic episode, followed at the long term by degeneration of 80% of synaptic contacts. In presynaptic terminals, ischemia induced a depletion of synaptic vesicles and changes in their spatial arrangement: they became more distant from active zones and had larger intervesicle spacing compared to controls. These rapid structural synaptic changes could be implicated in the mechanisms of cell death or adaptive plasticity. Comparison of the in vivo and in vitro model systems used in the study demonstrated a general similarity of these early morphological changes, confirming the validity of the in vitro model for studying synaptic structural plasticity.


Subject(s)
Hippocampus/pathology , Hypoxia-Ischemia, Brain/pathology , Nerve Degeneration/pathology , Synapses/pathology , Animals , Animals, Newborn , Cell Death/physiology , Disease Models, Animal , Excitatory Postsynaptic Potentials/physiology , Hippocampus/blood supply , Hippocampus/physiopathology , Hypoxia-Ischemia, Brain/physiopathology , Microscopy, Electron, Transmission , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Nerve Regeneration/physiology , Neural Pathways/blood supply , Neural Pathways/pathology , Neural Pathways/physiopathology , Neuronal Plasticity/physiology , Organ Culture Techniques , Presynaptic Terminals/pathology , Pyramidal Cells/pathology , Rats , Synaptic Membranes/pathology , Synaptic Transmission/physiology , Synaptic Vesicles/pathology
18.
Eur J Neurosci ; 22(7): 1589-96, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16197499

ABSTRACT

There is a major unmet need for development of innovative strategies for neuroprotection against ischemic brain injury. Here we show that FGL, a neural cell adhesion molecule (NCAM)-derived peptide binding to and inducing phosphorylation of the fibroblast growth factor receptor (FGFR), acts neuroprotectively after an ischemic insult both in vitro and in vivo. The neuroprotective activity of FGL was tested in vitro on dissociated rat hippocampal neurons and hippocampal slice cultures, using a protocol of oxygen-glucose deprivation (OGD). FGL protected hippocampal neurons from damage and maintained or restored their metabolic and presynaptic activity, both if employed as a pretreatment alone to OGD, and if only applied after the insult. In vivo 24 h pretreatment with a single suboccipital injection of FGL significantly protected hippocampal CA1 neurons from death in a transient global ischemia model in the gerbil. We conclude that FGL promotes neuronal survival after ischemic brain injury.


Subject(s)
Brain Ischemia/prevention & control , Hippocampus/cytology , Neural Cell Adhesion Molecules/administration & dosage , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Animals , Animals, Newborn , Cell Count/methods , Cells, Cultured , Drug Interactions , Glucose/deficiency , Hypoxia , Neural Cell Adhesion Molecules/chemical synthesis , Neuroprotective Agents/chemical synthesis , Organ Culture Techniques , Phosphorylation/drug effects , Propidium , Pyridinium Compounds/metabolism , Pyrroles/pharmacology , Quaternary Ammonium Compounds/metabolism , Rats , Rats, Wistar , Receptors, Fibroblast Growth Factor/metabolism , Synapses/pathology , Tetrazolium Salts/metabolism , Thiazoles/metabolism , Time Factors
19.
J Neurosci ; 24(17): 4197-204, 2004 Apr 28.
Article in English | MEDLINE | ID: mdl-15115815

ABSTRACT

The neural cell adhesion molecule (NCAM) plays a critical role in development and plasticity of the nervous system and is involved in the mechanisms of learning and memory. Here, we show that intracerebroventricular administration of the FG loop (FGL), a synthetic 15 amino acid peptide corresponding to the binding site of NCAM for the fibroblast growth factor receptor 1 (FGFR1), immediately after training rats in fear conditioning or water maze learning, induced a long-lasting improvement of memory. In primary cultures of hippocampal neurons, FGL enhanced the presynaptic function through activation of FGFR1 and promoted synapse formation. These results provide the first evidence for a memory-facilitating effect resulting from a treatment that mimics NCAM function. They suggest that increased efficacy of synaptic transmission and formation of new synapses probably mediate the cognition-enhancing properties displayed by the peptide.


Subject(s)
Memory/drug effects , Molecular Mimicry/physiology , Neural Cell Adhesion Molecules/agonists , Neural Cell Adhesion Molecules/pharmacology , Peptides/pharmacology , Synapses/drug effects , Synaptic Transmission/drug effects , Amino Acid Sequence , Animals , Cells, Cultured , Conditioning, Classical/drug effects , Emotions/drug effects , Injections, Intraventricular , Male , Maze Learning/drug effects , Memory/physiology , Molecular Sequence Data , Motor Activity/drug effects , Neural Cell Adhesion Molecules/chemistry , Neurons/drug effects , Neurons/physiology , Peptides/chemistry , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Pyrroles/pharmacology , Rats , Rats, Wistar , Receptors, Fibroblast Growth Factor/drug effects , Receptors, Fibroblast Growth Factor/metabolism , Synapses/physiology , Synaptic Transmission/physiology
20.
Microsc Res Tech ; 65(6): 287-91, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15662622

ABSTRACT

Synaptic vesicles are organelles that specialize in the storage of a neurotransmitter that continuously undergo an exo-endocytotic cycle. During this cycle vesicles change their positions within a presynaptic terminal and their numbers as well as spatial arrangement can provide insight into a neurotransmitter turnover. This article introduces a technique based on the nearest-neighbor formalism to quantify the proximity of vesicles to active zones and vesicle clustering in different regions of a terminal. The technique, implemented in a software package, uses the two-dimensional coordinates of features identified in digitized electron micrographs as an input. It has been validated in the analysis of asymmetric synapses of the rat hippocampal CA1 stratum radiatum affected by transient cerebral ischemia. It was shown that a 15-minute-long ischemic episode influenced the spatial arrangement of vesicles that were more distant from active zones and had larger intervesicle spacings with respect to the control. The latter effect was apparently stronger within 200 nm distance of active zones.


Subject(s)
Synaptic Vesicles/ultrastructure , Animals , Brain Ischemia/pathology , Hippocampus/ultrastructure , Male , Microscopy, Electron , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL