Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS One ; 8(12): e81043, 2013.
Article in English | MEDLINE | ID: mdl-24339894

ABSTRACT

The blood-brain barrier prevents the entry of many therapeutic agents into the brain. Various nanocarriers have been developed to help agents to cross this barrier, but they all have limitations, with regard to tissue-selectivity and their ability to cross the endothelium. This study investigated the potential for 4 nm coated gold nanoparticles to act as selective carriers across human brain endothelium and subsequently to enter astrocytes. The transfer rate of glucose-coated gold nanoparticles across primary human brain endothelium was at least three times faster than across non-brain endothelia. Movement of these nanoparticles occurred across the apical and basal plasma membranes via the cytosol with relatively little vesicular or paracellular migration; antibiotics that interfere with vesicular transport did not block migration. The transfer rate was also dependent on the surface coating of the nanoparticle and incubation temperature. Using a novel 3-dimensional co-culture system, which includes primary human astrocytes and a brain endothelial cell line hCMEC/D3, we demonstrated that the glucose-coated nanoparticles traverse the endothelium, move through the extracellular matrix and localize in astrocytes. The movement of the nanoparticles through the matrix was >10 µm/hour and they appeared in the nuclei of the astrocytes in considerable numbers. These nanoparticles have the correct properties for efficient and selective carriers of therapeutic agents across the blood-brain barrier.


Subject(s)
Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Glucose/chemistry , Gold/chemistry , Gold/metabolism , Metal Nanoparticles/chemistry , Biological Transport , Blood-Brain Barrier/metabolism , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Carriers/toxicity , Endothelium/metabolism , Gold/toxicity , Humans , Intracellular Space/metabolism
2.
PLoS One ; 8(8): e71479, 2013.
Article in English | MEDLINE | ID: mdl-23951173

ABSTRACT

Increased levels of neurotoxic amyloid-beta in the brain are a prominent feature of Alzheimer's disease. FG-Loop (FGL), a neural cell adhesion molecule-derived peptide that corresponds to its second fibronectin type III module, has been shown to provide neuroprotection against a range of cellular insults. In the present study impairments in social recognition memory were seen 24 days after a 5 mg/15 µl amyloid-beta(25-35) injection into the right lateral ventricle of the young adult rat brain. This impairment was prevented if the animal was given a systemic treatment of FGL. Unbiased stereology was used to investigate the ability of FGL to alleviate the deleterious effects on CA1 pyramidal cells of the amyloid-beta(25-35) injection. NeuN, a neuronal marker (for nuclear staining) was used to identify pyramidal cells, and immunocytochemistry was also used to identify inactive glycogen synthase kinase 3beta (GSK3ß) and to determine the effects of amyloid-beta(25-35) and FGL on the activation state of GSK3ß, since active GSK3ß has been shown to cause a range of AD pathologies. The cognitive deficits were not due to hippocampal atrophy as volume estimations of the entire hippocampus and its regions showed no significant loss, but amyloid-beta caused a 40% loss of pyramidal cells in the dorsal CA1 which was alleviated partially by FGL. However, FGL treatment without amyloid-beta was also found to cause a 40% decrease in CA1 pyramidal cells. The action of FGL may be due to inactivation of GSK3ß, as an increased proportion of CA1 pyramidal neurons contained inactive GSK3ß after FGL treatment. These data suggest that FGL, although potentially disruptive in non-pathological conditions, can be neuroprotective in disease-like conditions.


Subject(s)
Amyloid beta-Peptides/adverse effects , CA1 Region, Hippocampal/drug effects , Memory/drug effects , Neural Cell Adhesion Molecules/pharmacology , Peptide Fragments/adverse effects , Pyramidal Cells/drug effects , Amyloid beta-Peptides/administration & dosage , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Cell Count , Gene Expression/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Injections, Intravenous , Injections, Intraventricular , Male , Memory/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Peptide Fragments/administration & dosage , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Rats , Rats, Wistar
3.
Biomaterials ; 34(30): 7335-43, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23834895

ABSTRACT

A new combination of tissue engineering techniques provides a simple and effective method for building aligned cellular biomaterials. Self-alignment of Schwann cells within a tethered type-1 collagen matrix, followed by removal of interstitial fluid produces a stable tissue-like biomaterial that recreates the aligned cellular and extracellular matrix architecture associated with nerve grafts. Sheets of this engineered neural tissue supported and directed neuronal growth in a co-culture model, and initial in vivo tests showed that a device containing rods of rolled-up sheets could support neuronal growth during rat sciatic nerve repair (5 mm gap). Further testing of this device for repair of a critical-sized 15 mm gap showed that, at 8 weeks, engineered neural tissue had supported robust neuronal regeneration across the gap. This is, therefore, a useful new approach for generating anisotropic engineered tissues, and it can be used with Schwann cells to fabricate artificial neural tissue for peripheral nerve repair.


Subject(s)
Nerve Regeneration , Nerve Tissue/pathology , Nerve Tissue/physiopathology , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Tissue Engineering , Animals , Cell Proliferation , Cell Survival , Disease Models, Animal , Neurites/metabolism , Prosthesis Implantation , Rats , Rats, Sprague-Dawley , Schwann Cells/cytology , Schwann Cells/ultrastructure
4.
J Immunol ; 189(6): 3130-9, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22896632

ABSTRACT

During neuroinflammation, cytokines such as TNF-α and IFN-γ secreted by activated leukocytes and/or CNS resident cells have been shown to alter the phenotype and function of brain endothelial cells (BECs) leading to blood-brain barrier breakdown. In this study, we show that the human BEC line hCMEC/D3 expresses the receptors for TNF-α, TNF receptor 1 and TNF receptor 2, and for IFN-γ. BEC activation with TNF-α alone or in combination with IFN-γ induced endothelial leakage of paracellular tracers. At high cytokine concentrations (10 and 100 ng/ml), this effect was associated with caspase-3/7 activation and apoptotic cell death as evidenced by annexin V staining and DNA fragmentation (TUNEL) assays. In addition, inhibition of JNK and protein kinase C activation at these doses partially prevented activation of caspase-3/7, although only JNK inhibition was partially able to prevent the increase in BEC paracellular permeability induced by cytokines. By contrast, lower cytokine concentrations (1 ng/ml) also led to effector caspase activation, increased paracellular flux, and redistribution of zonula occludens-1 and VE-cadherin but failed to induce apoptosis. Under these conditions, specific caspase-3 and caspase-9, but not caspase-8, inhibitors partially blocked cytokine-induced disruption of tight and adherens junctions and BEC paracellular permeability. Our results suggest that the concentration of cytokines in the CNS endothelial microenvironment determines the extent of caspase-mediated barrier permeability changes, which may be generalized as a result of apoptosis or more subtle as a result of alterations in the organization of junctional complex molecules.


Subject(s)
Blood-Brain Barrier/enzymology , Blood-Brain Barrier/immunology , Brain/enzymology , Brain/immunology , Cytokines/physiology , Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Blood-Brain Barrier/pathology , Brain/pathology , Cell Line , Endothelium, Vascular/pathology , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interferon-gamma/metabolism , Microcirculation/immunology , Receptors, Interferon/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/metabolism , Subcellular Fractions/enzymology , Subcellular Fractions/immunology , Subcellular Fractions/pathology , Interferon gamma Receptor
5.
Proc Natl Acad Sci U S A ; 108(41): 17213-8, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21969592

ABSTRACT

Stress has been identified as a major causal factor for many mental disorders. However, our knowledge about the chain of molecular and cellular events translating stress experience into altered behavior is still rather scant. Here, we have characterized a murine ortholog of the putative tumor suppressor gene DRR1 as a unique stress-induced protein in brain. It binds to actin, promotes bundling and stabilization of actin filaments, and impacts on actin-dependent neurite outgrowth. Endogenous DRR1 localizes to some, but not all, synapses, with preference for the presynaptic region. Hippocampal virus-mediated enhancement of DRR1 expression reduced spine density, diminished the probability of synaptic glutamate release, and altered cognitive performance. DRR1 emerges as a protein to link stress with actin dynamics, which in addition is able to act on synaptic function and cognition.


Subject(s)
Cognition/physiology , Synapses/physiology , Tumor Suppressor Proteins/physiology , Actins/metabolism , Animals , Behavior, Animal/physiology , Brain/cytology , Brain/physiology , Genes, Tumor Suppressor , HEK293 Cells , Humans , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurites/metabolism , Neurites/ultrastructure , Protein Binding , Stress, Physiological , Tumor Suppressor Proteins/genetics
6.
Brain Res ; 1292: 14-24, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19631619

ABSTRACT

P-glycoprotein (P-gp) expression at the blood-brain barrier prevents unwanted blood-borne toxins and signalling molecules from entering the brain. Primary and immortalised human brain endothelial cells (BECs) represent two suitable options for studying P-gp function in vitro. The limited supply of primary human BECs and their instability over passage number make this choice unattractive for medium/high throughput studies. The aim of this study was to further characterise the expression of P-gp by an immortalised human BEC line, hCMEC/D3, in order to evaluate their use as an in vitro human blood-brain barrier model. P-gp expression was stable over a high passage number (up to passage 38) and was polarised on the apical plasma membrane, consistent with human BECs in vivo. In addition, hCMEC/D3 cell P-gp expression was comparable, albeit slightly lower to that observed in primary isolated human BECs although P-gp function was similar in both cell lines. The P-gp inhibitors tariquidar and vinblastine prevented the efflux of rhodamine 123 (rh123) from hCMEC/D3 cells, indicative of functional P-gp expression. hCMEC/D3 cells also displayed polarised P-gp transport, since both tariquidar and vinblasine selectively increased the apical-to-basolateral permeability of hCMEC/D3 cells to rh123. The results presented here demonstrate that hCMEC/D3 cells are a suitable model to investigate substrate specificity of P-gp in BECs of human origin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/physiology , Cell Membrane Permeability/physiology , Endothelial Cells/physiology , Fluorescent Dyes/pharmacokinetics , Rhodamine 123/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Biological Transport/drug effects , Blood-Brain Barrier/physiology , Brain/drug effects , Capillary Permeability , Cell Line , Cell Membrane/drug effects , Cell Membrane/physiology , Cells, Cultured , Central Nervous System Agents/pharmacology , Endothelial Cells/drug effects , Humans , Models, Biological , Quinolines/pharmacology , Vinblastine/pharmacology
7.
J Neuropathol Exp Neurol ; 68(3): 227-40, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19225413

ABSTRACT

Leukocyte migration into the central nervous system (CNS) is mediated by chemokines expressed on CNS endothelial cell surfaces. This study investigated the production of chemokines and expression of chemokine receptors by human brain endothelial cells (HBECs) in vitro and in situ. Four chemokines (CCL2, CCL5, CXCL8, and CXCL10) were demonstrated by immunohistochemistry in endothelial cells in brain samples from patients with multiple sclerosis. CXCL8 and CCL2 were constitutively released and increased by primary HBECs and the brain endothelial cell line hCEMC/D3 in response to tumor necrosis factor and/or interferon gamma. CXCL10 and CCL5 were undetectable in resting endothelial cells but were secreted in response to these proinflammatory cytokines. Tumor necrosis factor strongly increased the production of CCL2, CCL5, and CXCL8; interferon gamma upregulated CXCL10 exclusively. CCL3 was not secreted by HBECs and seemed to be confined to astrocytes in situ. The chemokine receptors CXCR1 and CXCR3 were expressed by HBECs both in vitro and in situ; CXCR3 was upregulated in response to cytokine stimulation in vitro. In contrast, CXCR3 expression was reduced in noninflammatory (silent) multiple sclerosis lesions. The particularly high levels of CXCL10 and CXCL8 expressed by brain endothelium may contribute to the predominant TH1-type inflammatory response observed in chronic inflammatory conditions such as multiple sclerosis.


Subject(s)
Brain/metabolism , Chemokines/biosynthesis , Endothelium, Vascular/metabolism , Multiple Sclerosis/metabolism , Receptors, Chemokine/biosynthesis , Adult , Aged , Aged, 80 and over , Brain/blood supply , Brain/immunology , Chemotaxis, Leukocyte , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Inflammation/immunology , Male , Microscopy, Electron, Transmission , Middle Aged , Multiple Sclerosis/immunology , Reverse Transcriptase Polymerase Chain Reaction
8.
Eur J Neurosci ; 27(2): 301-14, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18215229

ABSTRACT

The FGL peptide is a neural cell adhesion molecule (NCAM) mimetic comprising a 15-amino-acid-long sequence of the FG loop region of the second fibronectin type III module of NCAM. It corresponds to the binding site of NCAM for the fibroblast growth factor receptor 1. FGL improves cognitive function through enhancement of synaptic function. We examined the effect of FGL on synaptic and dendritic structure in the brains of aged (22-month-old) rats that were injected subcutaneously (8 mg/kg) at 2-day intervals until 19 days after the start of the experiment. Animals were perfused with fixative, brains removed and coronal sections cut at 50 microm. The hippocampal volume was measured, tissue embedded and ultrathin sections viewed in a JEOL 1010 electron microscope. Analyses were made of synaptic and dendritic parameters following three-dimensional reconstruction via images from a series of approximately 100 serial ultrathin sections. FGL affected neither hippocampal volume nor spine or synaptic density in the middle molecular layer of the dentate gyrus. However, it increased the ratio of mushroom to thin spines, number of multivesicular bodies and also increased the frequency of appearance of coated pits. Three-dimensional analysis showed a significant decrease in both post-synaptic density and apposition zone curvature of mushroom spines following FGL treatment, whereas for thin spines the convexity of the apposition zone increased. These data indicate that FGL induces large changes in the fine structure of synapses and dendritic spines in hippocampus of aged rats, complementing data showing its effect on cognitive processes.


Subject(s)
Aging , Biomimetic Materials/pharmacology , Dendritic Spines/ultrastructure , Dentate Gyrus/ultrastructure , Neural Cell Adhesion Molecules/pharmacology , Synapses/ultrastructure , Aging/drug effects , Animals , Cell Adhesion Molecules/pharmacology , Dendritic Spines/drug effects , Dentate Gyrus/drug effects , Imaging, Three-Dimensional/methods , Male , Rats , Rats, Wistar , Synapses/drug effects
9.
Eur J Neurosci ; 27(2): 315-25, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18215230

ABSTRACT

In adult brain the majority of AMPA glutamate receptor (GluR) subunits contain GluR2. In knock-out (KO) mice the absence of GluR2 results in consequences for synaptic plasticity including cognitive impairments. Here the morphology of dendritic spines and their synaptic contacts was analysed via three-dimensional reconstruction of serial electron micrographs from dentate gyrus (DG) of adult wild type (WT) and GluR2 KO mice. Pre-embedding immunocytochemical staining was used to examine the distribution and subcellular localization of AMPA receptor GluR1 and N-methyl-D-aspartate receptor NR1 subunits. There were no significant changes in synapse density in the DG of GluR2 KO compared with WT mice. However, in GluR2 KO mice there was a significant decrease in the percentage of synapses on mushroom spines but an increase in synapses on thin spines. There was also a large decrease in the proportion of synapses with complex perforated/segmented post-synaptic densities (PSDs) (25 vs. 78% in WT) but an increase in synapses with macular PSDs (75 vs. 22%). These data were coupled in GluR2 KO mice with significant decreases in volume and surface area of mushroom spines and their PSDs. In both GluR2 KO and WT mice, NR1 and GluR1 receptors were present in dendrites and spines but there was a significant reduction in NR1 labelling of spine membranes and cytoplasm in GluR2 KO mice, and a small decrease in GluR1 immunolabelling in membranes and cytoplasm of spines in GluR2 KO compared with WT mice. Our data demonstrate that the absence of GluR2 has a significant effect on both DG synapse and spine cytoarchitecture and the expression of NR1 receptors.


Subject(s)
Dendritic Spines/ultrastructure , Dentate Gyrus/ultrastructure , Receptors, AMPA/biosynthesis , Synapses/ultrastructure , Animals , Dendritic Spines/metabolism , Dentate Gyrus/metabolism , Gene Expression Regulation/physiology , Male , Mice , Mice, Knockout , Receptors, AMPA/genetics , Synapses/metabolism
10.
Neuron Glia Biol ; 4(3): 169-78, 2008 Aug.
Article in English | MEDLINE | ID: mdl-19674508

ABSTRACT

Investigations examining the role of polysialic acid (PSA) on the neural cell adhesion molecule (NCAM) in synaptic plasticity have yielded inconsistent data. Here, we addressed this issue by determining whether homosynaptic long-term potentiation (LTP) and heterosynaptic long-term depression (LTD) induce changes in the distribution of PSA-NCAM in the dentate gyrus (DG) of rats in vivo. In addition, we also examined whether the observed modifications were initiated via the activation of N-methyl-D-aspartate (NMDA) receptors. Immunocytochemical analysis showed an increase in PSA-NCAM positive cells both at 2 and 24 h following high-frequency stimulation of either medial or lateral perforant paths, leading to homosynaptic LTP and heterosynaptic LTD, respectively, in the medial molecular layer of the DG. Analysis of sub-cellular distribution of PSA-NCAM by electron microscopy showed decreased PSA dendritic labelling in LTD rats and a sub-cellular relocation towards the spines in LTP rats. Importantly, these modifications were found to be independent of the activation of NMDA receptors. Our findings suggest that strong activation of the granule cells up-regulates PSA-NCAM synthesis which then incorporates into activated synapses, representing NMDA-independent plastic processes that act synergistically on LTP/LTD mechanisms without participating in their expression.


Subject(s)
Dentate Gyrus/physiology , Long-Term Potentiation/physiology , Neural Cell Adhesion Molecule L1/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Sialic Acids/metabolism , Synapses/physiology , Wakefulness , Animals , Biophysics , Dendrites/metabolism , Dendrites/ultrastructure , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , Long-Term Potentiation/drug effects , Male , Microscopy, Electron, Transmission , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Synapses/classification , Synapses/ultrastructure , Time Factors
11.
Endothelium ; 14(1): 7-15, 2007.
Article in English | MEDLINE | ID: mdl-17364892

ABSTRACT

Leukocyte migration across vascular endothelium is mediated by chemokines that are either synthesized by the endothelium or transferred across the endothelium from the tissue. The mechanism of transfer of two chemokines, CXCL10 (interferon gamma-inducible protein [IP]-10) and CCL2 (macrophage chemotactic protein [MCP]-1), was compared across dermal and lung microvessel endothelium and saphenous vein endothelium. The rate of transfer depended on both the type of endothelium and the chemokine. The permeability coefficient (Pe) for CCL2 movement across saphenous vein was twice the value for dermal endothelium and four times that for lung endothelium. In contrast, the Pe value for CXCL10 was lower for saphenous vein endothelium than the other endothelia. The differences in transfer rate between endothelia was not related to variation in paracellular permeability using a paracellular tracer, inulin, and immunoelectron microscopy showed that CXCL10 was transferred from the basal membrane in a vesicular compartment, before distribution to the apical membrane. Although all three endothelia expressed high levels of the receptor for CXCL10 (CXCR3), the transfer was not readily saturable and did not appear to be receptor dependent. After 30 min, the chemokine started to be reinternalized from the apical membrane in clathrin-coated vesicles. The data suggest a model for chemokine transcytosis, with a separate pathway for clearance of the apical surface.


Subject(s)
Chemokine CCL2/metabolism , Chemokines, CXC/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Biological Transport , Cell Culture Techniques , Cell Membrane Permeability , Cells, Cultured , Chemokine CCL2/analysis , Chemokine CCL2/biosynthesis , Chemokine CXCL10 , Chemokines, CXC/analysis , Chemokines, CXC/biosynthesis , Electric Impedance , Endocytosis , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/ultrastructure , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Humans , Inulin/metabolism , Kinetics , Lung/blood supply , Lung/cytology , Receptors, CCR2 , Receptors, CXCR3 , Receptors, Chemokine/analysis , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Skin/blood supply , Skin/cytology
12.
J Neurochem ; 99(4): 1207-23, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17026528

ABSTRACT

The somatodendritic A-current, I(SA), in hippocampal CA1 pyramidal neurons regulates the processing of synaptic inputs and the amplitude of back propagating action potentials into the dendritic tree, as well as the action potential firing properties at the soma. In this study, we have used RNA interference and over-expression to show that expression of the Kv4.2 gene specifically regulates the I(SA) component of A-current in these neurons. In dissociated hippocampal pyramidal neuron cultures, or organotypic cultured CA1 pyramidal neurons, the expression level of Kv4.2 is such that the I(SA) channels are maintained in the population at a peak conductance of approximately 950 pS/pF. Suppression of Kv4.2 transcripts in hippocampal pyramidal neurons using an RNA interference vector suppresses I(SA) current by 60% in 2 days, similar to the effect of expressing dominant-negative Kv4 channel constructs. Increasing the expression of Kv4.2 in these neurons increases the level of I(SA) to 170% of the normal set point without altering the biophysical properties. Our results establish a specific role for native Kv4.2 transcripts in forming and maintaining I(SA) current at characteristic levels in hippocampal pyramidal neurons.


Subject(s)
Action Potentials/genetics , Hippocampus/metabolism , Pyramidal Cells/metabolism , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Action Potentials/drug effects , Animals , COS Cells , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cells, Cultured , Chlorocebus aethiops , Dendrites/metabolism , Down-Regulation/genetics , Gene Expression Regulation/genetics , Genetic Vectors/genetics , Hippocampus/drug effects , Hippocampus/ultrastructure , Microscopy, Electron, Transmission , Organ Culture Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/ultrastructure , RNA Interference , Rats , Rats, Sprague-Dawley , Shal Potassium Channels/drug effects , Synaptic Transmission/genetics
13.
J Comp Neurol ; 492(1): 50-65, 2005 Nov 07.
Article in English | MEDLINE | ID: mdl-16175555

ABSTRACT

The fine structure of mitochondria and smooth endoplasmic reticulum (SER) was studied via electron microscopy in dendritic and axonal neuronal segments of hippocampal areas CA1, CA3, and dentate gyrus (DG) of both ground squirrels in normothermic and hibernating conditions, and rats. Ultrathin serial sections of approximately 60 nm (up to 150 per series) were taken and three-dimensional (3D) reconstructions made of dendritic segments, up to 36 microm in length. Mitochondria were demonstrated to be present in filamentous form in every dendrite examined, in each of the hippocampal regions studied, whether in rat or ground squirrel. In addition, apparent continuity between the outer mitochondrial membrane and that of SER was observed by 3D reconstructions of very ultrathin (20 nm) serial sections prepared from dendritic segments. It is believed that SER penetrate into the heads of thin and mushroom spines but mitochondria do not enter the heads of these types of spines in dentate gyrus or CA1 of either rat or ground squirrel. However, in CA3 we have shown here that mitochondria penetrate into the base of the large thorny excrescences. Mushroom dendritic spines (but not thin spines) contained puncta adherentia, formed between pre- and postsynaptic membranes. In contrast to dendrites, the mitochondrial population of axonal processes in the same hippocampal regions were found only in the form of discrete bodies no more than 3 microm in length. The issue of the likely function of this network in dendrites and its potential role in calcium movement is discussed.


Subject(s)
Axons/ultrastructure , Dendrites/ultrastructure , Endoplasmic Reticulum, Smooth/ultrastructure , Intracellular Membranes/ultrastructure , Mitochondria/ultrastructure , Animals , Dendrites/classification , Dendritic Spines/ultrastructure , Female , Hibernation , Imaging, Three-Dimensional , Male , Rats , Rats, Sprague-Dawley , Sciuridae
14.
Eur J Neurosci ; 17(11): 2447-56, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12814376

ABSTRACT

The impact was examined of exposing rats to two life experiences of a very different nature (stress and learning) on synaptic structures in hippocampal area CA3. Rats were subjected to either (i) chronic restraint stress for 21 days, and/or (ii) spatial training in a Morris water maze. At the behavioural level, restraint stress induced an impairment of acquisition of the spatial response. Moreover, restraint stress and water maze training had contrasting impacts on CA3 synaptic morphometry. Chronic stress induced a loss of simple asymmetric synapses [those with an unperforated postsynaptic density (PSD)], whilst water maze learning reversed this effect, promoting a rapid recovery of stress-induced synaptic loss within 2-3 days following stress. In addition, in unstressed animals a correlation was found between learning efficiency and the density of synapses with an unperforated PSD: the better the performance in the water maze, the lower the synaptic density. Water maze training increased the number of perforated synapses (those with a segmented PSD) in CA3, both in stressed and, more notably, in unstressed rats. The distinct effects of stress and learning on CA3 synapses reported here provide a neuroanatomical basis for the reported divergent effects of these experiences on hippocampal synaptic activity, i.e. stress as a suppressor and learning as a promoter of synaptic plasticity.


Subject(s)
Hippocampus/physiology , Maze Learning/physiology , Recovery of Function/physiology , Stress, Physiological/physiopathology , Synapses/physiology , Animals , Behavior, Animal , Hippocampus/ultrastructure , Male , Microscopy, Electron , Rats , Rats, Wistar , Restraint, Physical/methods , Reversal Learning , Spatial Behavior , Synapses/ultrastructure , Time Factors
15.
Perspect Med Virol ; 3: 173-183, 1987.
Article in English | MEDLINE | ID: mdl-32287596

ABSTRACT

Coronaviruses are spherical, lipid-containing, enveloped particles with tear-dropshaped surface projections or peplomers. The genome is one molecule of ssRNA and the virions characteristically contain three major structural protein classes. The antigenic relationships of coronaviruses present a complex pattern. The geographic distribution of many coronaviruses is worldwide. Biological vectors of coronaviruses have not been reported, and the natural hosts form the major reservoirs for further infection. Coronavirus particles contain three major protein classes, within which the polypeptides vary in number and molecular weight between species. The apparent size and shapes of coronaviruses can vary considerably. Coronavirus particles are spherical, although negatively stained air-dried particles are often pleomorphic. The morphology of coronavirus surface projections can vary considerably between different strains. The conventional structure on negative staining consists of tear-drop-shaped projections, although cone-shaped projections are also observed. In all these cases, the projections have the same length of about 20 nm. Other coronaviruses have short as well as 20-nm projections. Projections with blebs on thin stalks have been reported for other coronaviruses.

SELECTION OF CITATIONS
SEARCH DETAIL
...