Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Brain Behav Immun ; 107: 419-431, 2023 01.
Article in English | MEDLINE | ID: mdl-35907582

ABSTRACT

Adolescent alcohol use can permanently alter brain function and lead to poor health outcomes in adulthood. Emerging evidence suggests that alcohol use can predispose individuals to pain disorders or exacerbate existing pain conditions, but the underlying neural mechanisms are currently unknown. Here we report that mice exposed to adolescent intermittent access to ethanol (AIE) exhibit increased pain sensitivity and depressive-like behaviors that persist for several weeks after alcohol cessation and are accompanied by elevated CD68 expression in microglia and reduced numbers of serotonin (5-HT)-expressing neurons in the dorsal raphe nucleus (DRN). 5-HT expression was also reduced in the thalamus, anterior cingulate cortex (ACC) and amygdala as well as the lumbar dorsal horn of the spinal cord. We further demonstrate that chronic minocycline administration after AIE alleviated hyperalgesia and social deficits, while chemogenetic activation of microglia in the DRN of ethanol-naïve mice reproduced the effects of AIE on pain and social behavior. Chemogenetic activation of microglia also reduced tryptophan hydroxylase 2 (Tph2) expression and was negatively correlated with the number of 5-HT-immunoreactive cells in the DRN. Taken together, these results indicate that microglial activation in the DRN may be a primary driver of pain, negative affect, and 5-HT depletion after AIE.


Subject(s)
Underage Drinking , Mice , Animals , Ethanol , Serotonin , Pain
2.
J Psychiatr Res ; 133: 205-211, 2021 01.
Article in English | MEDLINE | ID: mdl-33360427

ABSTRACT

Most of the animal studies using inflammation-induced cognitive change have relied on behavioral testing without objective and biologically solid methods to quantify the severity of cognitive disturbances. We have developed a bispectral EEG (BSEEG) method using a novel algorithm in clinical study. This method effectively differentiates between patients with and without delirium, and predict long-term mortality. In the present study, we aimed to apply our bispectral EEG (BSEEG) method, which can detect patients with delirium, to a mouse model of delirium with systemic inflammation induced by lipopolysaccharides (LPS) injection. We recorded EEG after LPS injection using wildtype early adulthood mice (2~3-month-old) and aged mice (18-19-month-old). Animal EEG recordings were converted for power spectral density to calculate BSEEG score using the similar BSEEG algorithm previously developed for our human study. The BSEEG score was relatively stable and slightly high during the day. Alternatively, the BSEEG score was erratic and low in average during the night. LPS injection increased the BSEEG score dose-dependently and diminished the diurnal changes. The mean BSEEG score increased much more in the aged mice group as dosage increased. Our results suggest that BSEEG method can objectively "quantify" level of neuro-Inflammation induced by systemic inflammation (LPS), and that this BSEEG method can be useful as a model of delirium in mice.


Subject(s)
Delirium , Animals , Disease Models, Animal , Electroencephalography , Humans , Inflammation/chemically induced , Lipopolysaccharides , Mice
3.
Brain Behav Immun ; 55: 49-59, 2016 07.
Article in English | MEDLINE | ID: mdl-26576723

ABSTRACT

During CNS development, microglia transform from highly mobile amoeboid-like cells to primitive ramified forms and, finally, to highly branched but relatively stationary cells in maturity. The factors that control developmental changes in microglia are largely unknown. Because microglia detect and clear apoptotic cells, developmental changes in microglia may be controlled by neuronal apoptosis. Here, we assessed the extent to which microglial cell density, morphology, motility, and migration are regulated by developmental apoptosis, focusing on the first postnatal week in the mouse hippocampus when the density of apoptotic bodies peaks at postnatal day 4 and declines sharply thereafter. Analysis of microglial form and distribution in situ over the first postnatal week showed that, although there was little change in the number of primary microglial branches, microglial cell density increased significantly, and microglia were often seen near or engulfing apoptotic bodies. Time-lapse imaging in hippocampal slices harvested at different times over the first postnatal week showed differences in microglial motility and migration that correlated with the density of apoptotic bodies. The extent to which these changes in microglia are driven by developmental neuronal apoptosis was assessed in tissues from BAX null mice lacking apoptosis. We found that apoptosis can lead to local microglial accumulation near apoptotic neurons in the pyramidal cell body layer but, unexpectedly, loss of apoptosis did not alter overall microglial cell density in vivo or microglial motility and migration in ex vivo tissue slices. These results demonstrate that developmental changes in microglial form, distribution, motility, and migration occur essentially normally in the absence of developmental apoptosis, indicating that factors other than neuronal apoptosis regulate these features of microglial development.


Subject(s)
Apoptosis/physiology , Cell Movement/physiology , Hippocampus/growth & development , Microglia/physiology , Animals , Animals, Newborn , Hippocampus/cytology , Mice , Microglia/cytology
4.
Arterioscler Thromb Vasc Biol ; 35(11): 2391-400, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26427793

ABSTRACT

OBJECTIVE: Cellular fibronectin containing extra domain A (EDA(+)-FN) is abundant in the arteries of patients with atherosclerosis. Several in vitro studies suggest that EDA(+)-FN interacts with Toll-like receptor 4 (TLR4). We tested the hypothesis that EDA(+)-FN exacerbates atherosclerosis through TLR4 in a clinically relevant model of atherosclerosis, the apolipoprotein E-deficient (Apoe(-/-)) mouse. APPROACH AND RESULTS: The extent of atherosclerosis was evaluated in whole aortae and cross sections of the aortic sinus in male and female EDA(-/-)Apoe(-/-) mice (which lack EDA(+)-FN), EDA(fl/fl)Apoe(-/-) mice (which constitutively express EDA(+)-FN), and control Apoe(-/-) mice fed a high-fat Western diet for 14 weeks. Irrespective of sex, EDA(fl/fl)Apoe(-/-) mice exhibited a 2-fold increase in atherosclerotic lesions (aorta and aortic sinus) and macrophage content within plaques, whereas EDA(-/-)Apoe(-/-) mice exhibited reduced atherosclerotic lesions (P<0.05 versus Apoe(-/-), n=10-12 mice/group), although cholesterol and triglyceride levels and circulating leukocytes were similar. Genetic ablation of TLR4 partially reversed atherosclerosis exacerbation in EDA(fl/fl)Apoe(-/-) mice (P<0.05) but had no effect on atherosclerotic lesions in EDA(-/-)Apoe(-/-) mice. Purified cellular FN, which contains EDA, potentiated dose-dependent NFκB-mediated inflammation (increased phospho-NFκB p65/NFκB p65, tumor necrosis factor-α, and interleukin-1ß) in bone marrow-derived macrophages from EDA(-/-)Apoe(-/-) mice but not from EDA(-/-)TLR4(-/-)Apoe(-/-) mice. Finally, using immunohistochemistry, we provide evidence for the first time that EDA(+)-FN colocalizes with macrophage TLR4 in murine aortic lesions and human coronary artery atherosclerotic plaques. CONCLUSIONS: Our findings reveal that TLR4 signaling contributes to EDA(+)-FN-mediated exacerbation of atherosclerosis. We suggest that EDA(+)-FN could be a therapeutic target in atherosclerosis.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Fibronectins/metabolism , Macrophages/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Coronary Vessels/metabolism , Coronary Vessels/pathology , Diet, High-Fat , Disease Models, Animal , Female , Fibronectins/deficiency , Fibronectins/genetics , Humans , Lipoproteins, LDL/metabolism , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Plaque, Atherosclerotic , Protein Isoforms , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
5.
Glia ; 63(10): 1694-713, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25856413

ABSTRACT

Fetal alcohol exposure is the most common known cause of preventable mental retardation, yet we know little about how microglia respond to, or are affected by, alcohol in the developing brain in vivo. Using an acute (single day) model of moderate (3 g/kg) to severe (5 g/kg) alcohol exposure in postnatal day (P) 7 or P8 mice, we found that alcohol-induced neuroapoptosis in the neocortex is closely correlated in space and time with the appearance of activated microglia near dead cells. The timing and molecular pattern of microglial activation varied with the level of cell death. Although microglia rapidly mobilized to contact and engulf late-stage apoptotic neurons, apoptotic bodies temporarily accumulated in neocortex, suggesting that in severe cases of alcohol toxicity the neurodegeneration rate exceeds the clearance capacity of endogenous microglia. Nevertheless, most dead cells were cleared and microglia began to deactivate within 1-2 days of the initial insult. Coincident with microglial activation and deactivation, there was a transient increase in expression of pro-inflammatory factors, TNFα and IL-1ß, after severe (5 g/kg) but not moderate (3 g/kg) EtOH levels. Alcohol-induced microglial activation and pro-inflammatory factor expression were largely abolished in BAX null mice lacking neuroapoptosis, indicating that microglial activation is primarily triggered by apoptosis rather than the alcohol. Therefore, acute alcohol exposure in the developing neocortex causes transient microglial activation and mobilization, promoting clearance of dead cells and tissue recovery. Moreover, cortical microglia show a remarkable capacity to rapidly deactivate following even severe neurodegenerative insults in the developing brain.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Microglia/drug effects , Neocortex , Nerve Degeneration , bcl-2-Associated X Protein/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/genetics , CD18 Antigens/metabolism , CX3C Chemokine Receptor 1 , Caspase 3/metabolism , Cell Count , Cytokines/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/drug effects , Neocortex/growth & development , Neocortex/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Peptides/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Receptors, Purinergic P2Y12/metabolism , Time Factors , bcl-2-Associated X Protein/genetics
6.
Cold Spring Harb Protoc ; 2013(12): 1142-8, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24298036

ABSTRACT

Here we describe a method for imaging fluorescently labeled parenchymal microglia (MG) in excised neonatal or adult rodent brain tissue slices. Using multichannel confocal or two-photon time-lapse imaging, the approach affords real-time analyses of MG behaviors, including motility, migration, chemotaxis, proliferation, and phagocytosis in live brain tissues. The method is applicable to acutely prepared tissue slices from developing and adult rodents and to slice cultures derived from neonatal rodents, including transgenic and green fluorescent protein reporter mice. A variety of fluorescent tags can be used to study the structure and physiology of MG in these preparations. Moreover, bath application of reagents (such as ATP) can establish spatial and temporal gradients that induce chemokinesis- and chemotaxis-like MG migration in tissue slices. Thus, the approach is useful for dissecting the molecular basis of MG behaviors and testing whether candidate reagents alter MG behavior and function in semi-intact central nervous system tissue preparations.


Subject(s)
Brain/cytology , Brain/physiology , Microglia/cytology , Microglia/physiology , Microscopy, Fluorescence, Multiphoton/methods , Optical Imaging/methods , Time-Lapse Imaging/methods , Animals , Genes, Reporter , Mice , Mice, Transgenic , Staining and Labeling/methods
7.
Neuropharmacology ; 73: 311-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23770338

ABSTRACT

Brain-resident microglia may promote tissue repair following stroke but, like other cells, they are vulnerable to ischemia. Here we identify mechanisms involved in microglial ischemic vulnerability. Using time-lapse imaging of cultured BV2 microglia, we show that simulated ischemia (oxygen-glucose deprivation; OGD) induces BV2 microglial cell death. Removal of extracellular Ca(2+) or application of Brilliant Blue G (BBG), a potent P2X7 receptor (P2X7R) antagonist, protected BV2 microglia from death. To validate and extend these in vitro findings, we assessed parenchymal microglia in freshly isolated hippocampal tissue slices from GFP-reporter mice (CX3CR1(GFP/+)). We confirmed that calcium removal or application of apyrase, an ATP-degrading enzyme, abolished OGD-induced microglial cell death in situ, consistent with involvement of ionotropic purinergic receptors. Indeed, whole cell recordings identified P2X7R-like currents in tissue microglia, and OGD-induced microglial cell death was inhibited by BBG. These pharmacological results were complemented by studies in tissue slices from P2X7R null mice, in which OGD-induced microglia cell death was reduced by nearly half. Together, these results indicate that stroke-like conditions induce calcium-dependent microglial cell death that is mediated in part by P2X7R. This is the first identification of a purinergic receptor regulating microglial survival in living brain tissues. From a therapeutic standpoint, these findings could help direct novel approaches to enhance microglial survival and function following stroke and other neuropathological conditions.


Subject(s)
Glucose/deficiency , Hypoxia/metabolism , Ischemia/metabolism , Microglia/cytology , Microglia/metabolism , Receptors, Purinergic P2X7/physiology , Animals , Apyrase/pharmacology , Calcium/metabolism , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Extracellular Space/metabolism , Female , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , Microglia/drug effects , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/genetics , Rosaniline Dyes/pharmacology
8.
J Neuroimmune Pharmacol ; 8(3): 494-509, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23354784

ABSTRACT

A century after Cajal identified a "third element" of the nervous system, many issues have been clarified about the identity and function of one of its major components, the microglia. Here, we review recent findings by microgliologists, highlighting results from imaging studies that are helping provide new views of microglial behavior and function. In vivo imaging in the intact adult rodent CNS has revolutionized our understanding of microglial behaviors in situ and has raised speculation about their function in the uninjured adult brain. Imaging studies in ex vivo mammalian tissue preparations and in intact model organisms including zebrafish are providing insights into microglial behaviors during brain development. These data suggest that microglia play important developmental roles in synapse remodeling, developmental apoptosis, phagocytic clearance, and angiogenesis. Because microglia also contribute to pathology, including neurodevelopmental and neurobehavioral disorders, ischemic injury, and neuropathic pain, promising new results raise the possibility of leveraging microglia for therapeutic roles. Finally, exciting recent work is addressing unanswered questions regarding the nature of microglial-neuronal communication. While it is now apparent that microglia play diverse roles in neural development, behavior, and pathology, future research using neuroimaging techniques will be essential to more fully exploit these intriguing cellular targets for effective therapeutic intervention applied to a variety of conditions.


Subject(s)
Brain/embryology , Microglia/physiology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Animals , Brain/pathology , Brain/physiology , Brain Ischemia/pathology , Cell Movement/physiology , Humans , Microglia/pathology , Stroke/pathology
9.
Glia ; 60(11): 1747-60, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22847985

ABSTRACT

As brain-resident immune cells, microglia (MG) survey the brain parenchyma to maintain homeostasis during development and following injury. Research in perinatal stroke, a leading cause of lifelong disability, has implicated MG as targets for therapeutic intervention during stroke. Although MG responses are complex, work in developing rodents suggests that MG limit brain damage after stroke. However, little is known about how energy-limiting conditions affect MG survival and mobility (motility and migration) in developing brain tissues. Here, we used confocal time-lapse imaging to monitor MG viability and mobility during hypoxia or oxygen-glucose deprivation (OGD) in hippocampal tissue slices derived from neonatal GFP-reporter mice (CX3CR1(GFP/+) ). We found that MG remain viable for at least 6 h of hypoxia but begin to die after 2 h of OGD, while both hypoxia and OGD reduce MG motility. Unexpectedly, some MG retain or recover motility during OGD and can engulf dead cells. Additionally, MG from younger neonates (P2-P3) are more resistant to OGD than those from older ones (P6-P7), indicating increasing vulnerability with developmental age. Finally, transient (2 h) OGD also increases MG death, and although motility is rapidly restored after transient OGD, it remains below control levels for many hours. Together, these results show that MG in neonatal mouse brain tissues are vulnerable to both transient and sustained OGD, and many MG die within hours after onset of OGD. Preventing MG death may, therefore, provide a strategy for promoting tissue restoration after stroke.


Subject(s)
Cell Movement/physiology , Cell Survival/physiology , Glucose/deficiency , Hippocampus/pathology , Hypoxia, Brain/pathology , Microglia/pathology , Animals , Cell Death/physiology , Hippocampus/physiopathology , Hypoxia, Brain/physiopathology , Mice , Microglia/physiology , Neurons/pathology , Neurons/physiology
10.
Glia ; 60(2): 175-88, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22052455

ABSTRACT

Glutamate transporters (GluTs) maintain a low ambient level of glutamate in the central nervous system (CNS) and shape the activation of glutamate receptors at synapses. Nevertheless, the mechanisms that regulate the trafficking and localization of transporters near sites of glutamate release are poorly understood. Here, we examined the subcellular distribution and dynamic remodeling of the predominant GluT GLT-1 (excitatory amino acid transporter 2, EAAT2) in developing hippocampal astrocytes. Immunolabeling revealed that endogenous GLT-1 is concentrated into discrete clusters along branches of developing astrocytes that were apposed preferentially to synapsin-1 positive synapses. Green fluorescent protein (GFP)-GLT-1 fusion proteins expressed in astrocytes also formed distinct clusters that lined the edges of astrocyte processes, as well as the tips of filopodia and spine-like structures. Time-lapse three-dimensional confocal imaging in tissue slices revealed that GFP-GLT-1 clusters were dynamically remodeled on a timescale of minutes. Some transporter clusters moved within developing astrocyte branches as filopodia extended and retracted, while others maintained stable positions at the tips of spine-like structures. Blockade of neuronal activity with tetrodotoxin reduced both the density and perisynaptic localization of GLT-1 clusters. Conversely, enhancement of neuronal activity increased the size of GLT-1 clusters and their proximity to synapses. Together, these findings indicate that neuronal activity influences both the organization of GluTs in developing astrocyte membranes and their position relative to synapses.


Subject(s)
Astrocytes/metabolism , Cell Differentiation/physiology , Excitatory Amino Acid Transporter 2/metabolism , Hippocampus/growth & development , Neurons/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Hippocampus/cytology , Neurons/cytology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Synapses/metabolism
11.
J Comp Neurol ; 519(16): 3327-45, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21800311

ABSTRACT

The rat auditory cortex is organized as a tonotopic map of sound frequency. This map is broadly tuned at birth and is refined during the first 3 weeks postnatal. The structural correlates underlying tonotopic map maturation and reorganization during development are poorly understood. We employed fluorescent dye ballistic labeling ("DiOlistics") alone, or in conjunction with immunohistochemistry, to quantify synaptogenesis in the auditory cortex of normal hearing rats. We show that the developmental appearance of dendritic protrusions, which include both immature filopodia and mature spines, on layers 2/3, 4, and 5 pyramidal and layer 4 spiny nonpyramidal neurons occurs in three phases: slow addition of dendritic protrusions from postnatal day 4 (P4) to P9, rapid addition of dendritic protrusions from P9 to P19, and a final phase where mature protrusion density is achieved (>P21). Next, we combined DiOlistics with immunohistochemical labeling of bassoon, a presynaptic scaffolding protein, as a novel method to categorize dendritic protrusions as either filopodia or mature spines in cortex fixed in vivo. Using this method we observed an increase in the spine-to-filopodium ratio from P9-P16, indicating a period of rapid spine maturation. Previous studies report mature spines as being shorter in length compared to filopodia. We similarly observed a reduction in protrusion length between P9 and P16, corroborating our immunohistochemical spine maturation data. These studies show that dendritic protrusion formation and spine maturation occur rapidly at a time previously shown to correspond to auditory cortical tonotopic map refinement (P11-P14), providing a structural correlate of physiological maturation.


Subject(s)
Auditory Cortex/growth & development , Dendritic Spines/ultrastructure , Neurogenesis , Animals , Female , Fluorescent Antibody Technique , Male , Microscopy, Confocal , Rats , Rats, Sprague-Dawley
12.
J Biol Chem ; 286(30): 26496-506, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21652711

ABSTRACT

The A kinase anchor protein AKAP150 recruits the cAMP-dependent protein kinase (PKA) to dendritic spines. Here we show that in AKAP150 (AKAP5) knock-out (KO) mice frequency of miniature excitatory post-synaptic currents (mEPSC) and inhibitory post-synaptic currents (mIPSC) are elevated at 2 weeks and, more modestly, 4 weeks of age in the hippocampal CA1 area versus litter mate WT mice. Linear spine density and ratio of AMPAR to NMDAR EPSC amplitudes were also increased. Amplitude and decay time of mEPSCs, decay time of mIPSCs, and spine size were unaltered. Mice in which the PKA anchoring C-terminal 36 residues of AKAP150 are deleted (D36) showed similar changes. Furthermore, whereas acute stimulation of PKA (2-4 h) increases spine density, prolonged PKA stimulation (48 h) reduces spine density in apical dendrites of CA1 pyramidal neurons in organotypic slice cultures. The data from the AKAP150 mutant mice show that AKAP150-anchored PKA chronically limits the number of spines with functional AMPARs at 2-4 weeks of age. However, synaptic transmission and spine density was normal at 8 weeks in KO and D36 mice. Thus AKAP150-independent mechanisms correct the aberrantly high number of active spines in juvenile AKAP150 KO and D36 mice during development.


Subject(s)
A Kinase Anchor Proteins/metabolism , Aging/physiology , Dendrites/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , A Kinase Anchor Proteins/genetics , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Hippocampus/metabolism , Inhibitory Postsynaptic Potentials/physiology , Male , Mice , Mice, Knockout , Pyramidal Cells/cytology , Pyramidal Cells/metabolism
14.
Biocell ; 33(2): 71-80, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19886034

ABSTRACT

The localization of GluR1 subunits of ionotropic glutamate receptors in the glial cells and inhibitory neurons of cerebellar cortex and their association with the climbing and parallel fibers, and basket cell axons were studied. Samples of P14 and P21 rat cerebellar cortex were exposed to a specific antibody against GluR1 subunit(s) ofAMPA receptors and were examined with confocal laser scanning microscopy. GluR1 strong immunoreactivity was confined to Purkinje cell and the molecular layer. Weak GluR1 immunoreactivity was observed surrounding some Golgi cells in the granule cell layer. Intense GluR1 immunoreactivity was localized around Purkinje, basket, and stellate cells. Purkinje cells expressed strong GluR1 immunoreactivity surrounding the cell body, primary dendritic trunk and secondary and tertiary spiny dendritic branches. Marked immunofluorescent staining was also detected in the Bergmann glial fibers at the level of middle and outer third molecular layer. Positive immunofluorescence staining was also observed surrounding basket and stellate cells, and in the capillary wall. These findings suggest the specific localization of GluR1 subunits ofAMPA receptors in Bergmann glial cells, inhibitory cerebellar neurons, and the associated excitatory glutamatergic circuits formed by climbing and parallel fibers, and by the inhibitory basket cell axons.


Subject(s)
Cerebellum/cytology , Neurons/metabolism , Protein Subunits/metabolism , Receptors, AMPA/metabolism , Animals , Calbindins , Disks Large Homolog 4 Protein , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neurons/cytology , Purkinje Cells/cytology , Purkinje Cells/metabolism , Rats , Rats, Wistar , S100 Calcium Binding Protein G/metabolism , Synapsins/metabolism
15.
Biocell ; 33(2): 71-80, Aug. 2009. ilus
Article in English | LILACS | ID: lil-595031

ABSTRACT

The localization of GluR1 subunits of ionotropic glutamate receptors in the glial cells and inhibitory neurons of cerebellar cortex and their association with the climbing and parallel fibers, and basket cell axons were studied. Samples of P14 and P21 rat cerebellar cortex were exposed to a specific antibody against GluR1 subunit(s) ofAMPA receptors and were examined with confocal laser scanning microscopy. GluR1 strong immunoreactivity was confined to Purkinje cell and the molecular layer. Weak GluR1 immunoreactivity was observed surrounding some Golgi cells in the granule cell layer. Intense GluR1 immunoreactivity was localized around Purkinje, basket, and stellate cells. Purkinje cells expressed strong GluR1 immunoreactivity surrounding the cell body, primary dendritic trunk and secondary and tertiary spiny den dritic branches. Marked immunofluorescent staining was also detected in the Bergmann glial fibers at the level of middle and outer third molecular layer. Positive immunofluorescence staining was also observed surrounding basket and stellate cells, and in the capillary wall. These findings suggest the specific localization of GluR1 subunits ofAMPA receptors in Bergmann glial cells, inhibitory cerebellar neurons, and the associated excitatory glutamatergic circuits formed by climbing and parallel fibers, and by the inhibitory basket cell axons.


Subject(s)
Animals , Rats , Purkinje Cells/cytology , Purkinje Cells/metabolism , Cerebellum/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , /metabolism , Neurons/cytology , Neurons/metabolism , Rats, Wistar , Receptors, AMPA/metabolism , Protein Subunits/metabolism
16.
Biocell ; 33(2): 71-80, Aug. 2009. ilus
Article in English | BINACIS | ID: bin-127212

ABSTRACT

The localization of GluR1 subunits of ionotropic glutamate receptors in the glial cells and inhibitory neurons of cerebellar cortex and their association with the climbing and parallel fibers, and basket cell axons were studied. Samples of P14 and P21 rat cerebellar cortex were exposed to a specific antibody against GluR1 subunit(s) ofAMPA receptors and were examined with confocal laser scanning microscopy. GluR1 strong immunoreactivity was confined to Purkinje cell and the molecular layer. Weak GluR1 immunoreactivity was observed surrounding some Golgi cells in the granule cell layer. Intense GluR1 immunoreactivity was localized around Purkinje, basket, and stellate cells. Purkinje cells expressed strong GluR1 immunoreactivity surrounding the cell body, primary dendritic trunk and secondary and tertiary spiny den dritic branches. Marked immunofluorescent staining was also detected in the Bergmann glial fibers at the level of middle and outer third molecular layer. Positive immunofluorescence staining was also observed surrounding basket and stellate cells, and in the capillary wall. These findings suggest the specific localization of GluR1 subunits ofAMPA receptors in Bergmann glial cells, inhibitory cerebellar neurons, and the associated excitatory glutamatergic circuits formed by climbing and parallel fibers, and by the inhibitory basket cell axons.(AU)


Subject(s)
Animals , Rats , S100 Calcium Binding Protein G/metabolism , Cerebellum/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Purkinje Cells/cytology , Purkinje Cells/metabolism , Neurons/cytology , Neurons/metabolism , Protein Subunits/metabolism , Rats, Wistar , Receptors, AMPA/metabolism
17.
J Neurosci Res ; 87(9): 1969-79, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19235894

ABSTRACT

Epileptiform activity (EA) in vivo and in vitro induces a loss of dendritic spines and synapses. Because CaMKII has been implicated in synaptogenesis and synaptic plasticity, we investigated the role of CaMKII in the effects of EA on spines, using rat hippocampal slice cultures. To visualize dendrites and postsynaptic densities (PSDs) in pyramidal neurons in the slices, we used biolistic transfection to express either free GFP or a PSD95-YFP construct that specifically labels PSDs. This allowed us to distinguish two classes of dendritic protrusions: spines that contain PSDs, and filopodia that lack PSDs and that are, on average, longer than spines. By these criteria, 48 hr of EA caused a decrease specifically in the number of spines. Immunoblots showed that EA increased CaMKII activity in the slices. Inhibition of CaMKII by expression of AIP, a specific peptide inhibitor of CaMKII, reduced spine number under basal conditions and failed to prevent EA-induced spine loss. However, under EA conditions, AIP increased the number of filopodia and the number of PSDs on the dendritic shaft. These data show at least two roles for CaMKII activity in maintenance and remodeling of dendritic spines under basal or EA conditions. First, CaMKII activity promotes the maintenance of spines and spine PSDs. Second, CaMKII activity suppresses EA-induced formation of filopodia and suppresses an increase in shaft PSDs, apparently by promoting translocation of PSDs from dendritic shafts to spines and/or selectively stabilizing spine rather than shaft PSDs.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/enzymology , Epilepsy/enzymology , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Animals , Animals, Newborn , Cerebral Cortex/enzymology , Cerebral Cortex/ultrastructure , Dendritic Spines/ultrastructure , Disks Large Homolog 4 Protein , Epilepsy/physiopathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Organ Culture Techniques , Protein Transport/physiology , Pseudopodia/enzymology , Pseudopodia/ultrastructure , Pyramidal Cells/enzymology , Pyramidal Cells/ultrastructure , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synaptic Membranes/enzymology , Synaptic Membranes/ultrastructure
18.
Glia ; 55(8): 873-84, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17405148

ABSTRACT

Traumatic CNS injury activates and mobilizes resident parenchymal microglia (MG), which rapidly accumulate near injured neurons where they transform into phagocytes. The mechanisms underlying this rapid 'homing' in situ are unknown. Using time-lapse confocal imaging in acutely excised neonatal hippocampal slices, we show that rapid accumulation of MG near somata of injured pyramidal neurons in the stratum pyramidale (SP) results from directed migration from tissue regions immediately adjacent to (<200 microm from) the SP. Time-lapse sequences also reveal a 'spreading activation wave' wherein MG situated progressively farther from the SP begin to migrate later and exhibit less directional migration toward the SP. Because purines have been implicated in MG activation and chemotaxis, we tested whether ATP/ADP released from injured pyramidal neurons might account for these patterns of MG behavior. Indeed, application of apyrase, which degrades extracellular ATP/ADP, inhibits MG motility and homing to injured neurons in the SP. Moreover, bath application of exogenous ATP/ADP disrupts MG homing by inducing directional migration toward the slice exterior and away from injured neurons. These results indicate that extracellular ATP/ADP is both necessary and sufficient to induce directional migration and rapid homing of neonatal MG to injured neurons in situ. Rapid, ATP/ADP-dependent MG homing may promote clearance of dead and dying cells and help limit secondary damage during the critical first few hours after neuronal injury.


Subject(s)
Cell Communication/immunology , Cell Movement/immunology , Hippocampus/immunology , Hippocampus/pathology , Microglia/immunology , Pyramidal Cells/pathology , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Apyrase/pharmacology , Brain Injuries/immunology , Brain Injuries/pathology , Cell Communication/drug effects , Cell Count , Cell Movement/drug effects , Hippocampus/growth & development , Mice , Mice, Inbred C57BL , Microglia/cytology , Microscopy, Confocal , Organ Culture Techniques , Phagocytosis/immunology , Pyramidal Cells/injuries , Rats , Rats, Sprague-Dawley
19.
CSH Protoc ; 2007: pdb.prot4848, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-21356949

ABSTRACT

INTRODUCTIONRodent organotypic hippocampal slice cultures (OHSCs) provide an outstanding preparation of central nervous system tissue for exploring the dynamic structural and physiological features of neuronal and glial cells within their native three-dimensional environments. It is a straightforward matter to obtain tissue slices from neonatal rodents. These slices culture well for periods up to several weeks and are easy to manipulate, allowing for a variety of in vitro experimental models. OHSCs provide good optical and physiological accessibility for studies involving live cell imaging, with high spatial and temporal resolution. This protocol is used to harvest tissues for both immunohistochemical labeling after fixation, and for confocal time-lapse imaging in live tissues labeled by a variety of fluorescent dyes or by biolistic or viral transfection.

20.
CSH Protoc ; 2007: pdb.prot4852, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-21356950

ABSTRACT

INTRODUCTIONThis protocol describes methods for fluorescence labeling and time-lapse confocal imaging of microglia in acutely prepared tissue slices from developing and adult animals, and to slice cultures derived from early post-natal day 7 (

SELECTION OF CITATIONS
SEARCH DETAIL
...