Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Sci Rep ; 9(1): 565, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679745

ABSTRACT

Following injury to the central nervous system, astrocytes perform critical and complex functions that both promote and antagonize neural repair. Understanding the molecular signaling pathways that coordinate their diverse functional properties is key to developing effective therapeutic strategies. In the healthy, adult CNS, Sonic hedgehog (Shh) signaling is active in mature, differentiated astrocytes. Shh has been shown to undergo injury-induced upregulation and promote neural repair. Here, we investigated whether Shh signaling mediates astrocyte response to injury. Surprisingly, we found that following an acute, focal injury, reactive astrocytes exhibit a pronounced reduction in Shh activity in a spatiotemporally-defined manner. Shh signaling is lost in reactive astrocytes at the lesion site, but persists in mild to moderately reactive astrocytes in distal tissues. Nevertheless, local pharmacological activation of the Shh pathway in astrocytes mitigates inflammation, consistent with a neuroprotective role for Shh signaling after injury. Interestingly, we find that Shh signaling is restored to baseline levels two weeks after injury, a time during which acute inflammation has largely subsided and lesions have matured. Taken together, these data suggest that endogenous Shh signaling in astrocytes is dynamically regulated in a context dependent manner. In addition, exogenous activation of the Shh pathway promotes neuroprotection mediated by reactive astrocytes.


Subject(s)
Astrocytes/metabolism , Head Injuries, Penetrating/metabolism , Hedgehog Proteins/metabolism , Neuroprotection/physiology , Prosencephalon/injuries , Animals , Cell Movement/drug effects , Cyclohexylamines/pharmacology , Female , Gene Expression Regulation , Gliosis/genetics , Hedgehog Proteins/genetics , Leukocytes/immunology , Male , Mice , Mice, Transgenic , Signal Transduction/drug effects , Smoothened Receptor/agonists , Smoothened Receptor/metabolism , Thiophenes/pharmacology , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
2.
Glia ; 66(10): 2158-2173, 2018 10.
Article in English | MEDLINE | ID: mdl-30194744

ABSTRACT

Microglia and blood-borne macrophages in injured or diseased brains are difficult to distinguish because they share many common characteristics. However, the identification of microglia-specific markers and the use of flow cytometry have recently made it easy to discriminate these types of cells. In this study, we analyzed the features of blood-borne macrophages, and activated and resting microglia in a rat traumatic brain injury (TBI) model. Oxidative injury was indicated in macrophages and neurons in TBI lesions by the presence of 8-hydroxy-2'-deoxyguanosine (8-OHdG). Generation of mitochondrial reactive oxygen species (ROS) was markedly observed in granulocytes and macrophages, but not in activated or resting microglia. Dihydroethidium staining supported microglia not being the major source of ROS in TBI lesions. Furthermore, macrophages expressed NADPH oxidase 2, interleukin-1ß (IL-1ß), and CD68 at higher levels than microglia. In contrast, microglia expressed transforming growth factor ß1 (TGFß1), interleukin-6 (IL-6), and tumor necrosis factor α at higher levels than macrophages. A hypnotic, bromovalerylurea (BU), which has anti-inflammatory effects, reduced both glycolysis and mitochondrial oxygen consumption. BU administration inhibited chemokine CCL2 expression, accumulation of monocytes/macrophages, 8-OHdG generation, mitochondrial ROS generation, and proinflammatory cytokine expression, and markedly ameliorated the outcome of the TBI model. Yet, BU did not inhibit microglial activation or expression of TGFß1 and insulin-like growth factor 1 (IGF-1). These results indicate that macrophages are the major aggravating cell type in TBI lesions, in particular during the acute phase. Activated microglia may even play favorable roles. Reduction of cellular energy metabolism in macrophages and suppression of CCL2 expression in injured tissue may lead to amelioration of TBI.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain Injuries, Traumatic/physiopathology , Bromisovalum/pharmacology , Hypnotics and Sedatives/pharmacology , Macrophages/physiology , Microglia/physiology , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Cells, Cultured , Chemokine CCL2/metabolism , Disease Models, Animal , Macrophages/drug effects , Male , Microglia/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Prosencephalon/drug effects , Prosencephalon/injuries , Prosencephalon/pathology , Prosencephalon/physiopathology , RNA, Messenger/metabolism , Rats, Wistar , Wounds, Stab/drug therapy , Wounds, Stab/pathology , Wounds, Stab/physiopathology
3.
Neurol Res ; 40(4): 318-323, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29473447

ABSTRACT

OBJECTIVE: Cerebral ischemia can trigger the ERK1/2 signaling cascade that enables the brain to adapt to ischemic injury. However, the mechanism of ERK1/2 in ischemic brain injury remains unclear. The aim of this study was to examine the roles of the ERK1/2 signaling pathway and NMDA receptors in the apoptosis of CA1 pyramidal neurons after ischemia/reperfusion (I/R). METHODS: Male Wistar rats were subjected to a sham or transient forebrain ischemia procedure. Animals received the intracerebroventricular injection of U0126 (5 µl, 0.2 µg/µl) or vehicle 30 min before ischemia. Homogenates of the hippocampal CA1 field were obtained from sham-operated and ischemic rats 6, 12 or 48 h after ischemia/reperfusion (n = 6 per group) and then subjected to Western blotting analysis and TUNEL staining. Caspase-3 activity was assayed with a colorimetric assay kit. RESULTS: We found that the phosphorylation level of ERK1/2 is increased in the CA1 region following transient I/R. Blocking the ERK1/2 signaling pathway by administration U0126 attenuated apoptotic neuronal cell death via inhibition of NMDA receptors. CONCLUSION: These findings suggest a novel mechanism by which the ERK1/2 signaling pathway affects the post-I/R apoptosis of CA1 pyramidal neurons, which will provide a therapeutic target for the treatment of stroke.


Subject(s)
Apoptosis , Brain Ischemia/complications , Butadienes/administration & dosage , CA1 Region, Hippocampal/drug effects , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Nitriles/administration & dosage , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Male , Neurons/metabolism , Phosphorylation , Prosencephalon/injuries , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
4.
Brain Res ; 1634: 45-56, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26764532

ABSTRACT

Slit2 is a secreted glycoprotein that was originally identified as a chemorepulsive factor in the developing brain; however, it was recently reported that Slit2 is associated with adult neuronal function including a variety of pathophysiological processes. To elucidate whether Slit2 is implicated in the pathophysiology of ischemic injury, we investigated the temporal changes and cellular localization of Slit2 and its predominant receptors, Robo1 and Robo4, for 28 days after transient forebrain ischemia. Slit2 and its receptors had similar overall expression patterns in the control and ischemic hippocampi. The ligand and receptors were constitutively expressed in hippocampal neurons in control animals; however, in animals with ischemic injury, their upregulation was detected in reactive astrocytes, but not in neurons or activated microglia, in the CA1 region. Astroglial induction of Slit2 and its receptors occurred by day 3 after reperfusion, and appeared to increase progressively until the final time point on day 28. Their temporal expression patterns overlapped with the time period in which reactive astrocytes undergo dynamic structural changes and appear hypertrophic in the ischemic hippocampus. The immunohistochemical data were consistent with the results of the immunoblot analyses, indicating that the expression of Slit2 and Robo increased progressively over the relatively long period of 28 days examined here. Collectively, these results suggest that Slit2/Robo signaling may be involved in regulating the astroglial reaction via autocrine or paracrine mechanisms in post-ischemic processes. Moreover, this may contribute to the dynamic morphological changes that occur in astrocytes in response to ischemic injury.


Subject(s)
Astrocytes/metabolism , Hippocampus/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Ischemic Attack, Transient/metabolism , Nerve Tissue Proteins/metabolism , Prosencephalon/injuries , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley , Time Factors , Roundabout Proteins
5.
Article in English | MEDLINE | ID: mdl-26319311

ABSTRACT

Auditory feedback (AF) plays a critical role in vocal learning. Previous studies in songbirds suggest that low-frequency (<~1 kHz) components may be salient cues in AF. We explored this with auditory stimuli including the bird's own song (BOS) and BOS variants with increased relative power at low frequencies (LBOS). We recorded single units from BOS-selective neurons in two forebrain nuclei (HVC and Area X) in anesthetized zebra finches. Song-evoked responses were analyzed based on both rate (spike counts) and temporal coding of spike trains. The BOS and LBOS tended to evoke similar spike-count responses in substantially overlapping populations of neurons in both HVC and Area X. Analysis of spike patterns demonstrated temporal coding information that discriminated among the BOS and LBOS stimuli significantly better than spike counts in the majority of HVC (94 %) and Area X (85 %) neurons. HVC neurons contained more and a broader range of temporal coding information to discriminate among the stimuli than Area X neurons. These results are consistent with a role of spike timing in coding differences in the spectral components of BOS in HVC and Area X neurons.


Subject(s)
Action Potentials/physiology , Auditory Perception/physiology , Evoked Potentials, Auditory/physiology , Feedback, Sensory/physiology , Neurons, Afferent/physiology , Prosencephalon/cytology , Acoustic Stimulation , Animals , Finches , Fourier Analysis , Prosencephalon/injuries , Prosencephalon/physiology , Vocalization, Animal/physiology
6.
J Vis Exp ; (100): e52825, 2015 Jun 29.
Article in English | MEDLINE | ID: mdl-26167674

ABSTRACT

Following injury to the CNS, astrocytes undergo a broad range of biochemical, morphological, and molecular changes collectively referred to as reactive astrogliosis. Reactive astrocytes exert both inflammatory and protective effects that inhibit and promote, respectively, neural repair. The mechanisms underlying the diverse functional properties of reactive astrogliosis are not well understood. Achieving a greater understanding of these mechanisms is critical to developing therapeutic strategies to treat the injured CNS. Here we demonstrate a method to trigger reactive astrogliosis in the adult mouse forebrain using a forebrain stab lesion. This lesion model is simple, reliable, and requires only a stereotaxic device and a scalpel blade to produce the injury. The use of stab lesions as an injury model in the forebrain is well established and amenable to studies addressing a broad range of neuropathological outcomes, such as neuronal degeneration, neuroinflammation, and disruptions in the blood brain barrier (BBB). Thus, the forebrain stab injury model serves as a powerful tool that can be applied for a broad range of studies on the CNS response to trauma.


Subject(s)
Brain Injuries/physiopathology , Gliosis/etiology , Prosencephalon/injuries , Prosencephalon/physiopathology , Animals , Astrocytes/pathology , Brain Injuries/pathology , Disease Models, Animal , Gliosis/physiopathology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Prosencephalon/pathology
7.
PLoS One ; 8(3): e59250, 2013.
Article in English | MEDLINE | ID: mdl-23555640

ABSTRACT

Traumatic brain injury (TBI) increases neurogenesis in the forebrain subventricular zone (SVZ) and the hippocampal dentate gyrus (DG). Transforming growth factor-ß (TGF-ß) superfamily cytokines are important regulators of adult neurogenesis, but their involvement in the regulation of this process after brain injury is unclear. We subjected adult mice to controlled cortical impact (CCI) injury, and isolated RNA from the SVZ and DG at different post-injury time points. qPCR array analysis showed that cortical injury caused significant alterations in the mRNA expression of components and targets of the TGF-ß, BMP, and activin signaling pathways in the SVZ and DG after injury, suggesting that these pathways could regulate post-injury neurogenesis. In both neurogenic regions, the injury also induced expression of Runt-related transcription factor-1 (Runx1), which can interact with intracellular TGF-ß Smad signaling pathways. CCI injury strongly induced Runx1 expression in activated and proliferating microglial cells throughout the neurogenic regions. Runx1 protein was also expressed in a subset of Nestin- and GFAP-expressing putative neural stem or progenitor cells in the DG and SVZ after injury. In the DG only, these Runx1+ progenitors proliferated. Our data suggest potential roles for Runx1 in the processes of microglial cell activation and proliferation and in neural stem cell proliferation after TBI.


Subject(s)
Brain Injuries/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Dentate Gyrus/metabolism , Gene Expression Regulation , Neurogenesis/genetics , Prosencephalon/metabolism , Transforming Growth Factor beta/genetics , Activins/genetics , Activins/metabolism , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/injuries , Glial Fibrillary Acidic Protein , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Mice , Microglia/cytology , Microglia/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Prosencephalon/cytology , Prosencephalon/injuries , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
8.
Nat Med ; 19(3): 351-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23435171

ABSTRACT

Previous attempts to identify neuroprotective targets by studying the ischemic cascade and devising ways to suppress it have failed to translate to efficacious therapies for acute ischemic stroke. We hypothesized that studying the molecular determinants of endogenous neuroprotection in two well-established paradigms, the resistance of CA3 hippocampal neurons to global ischemia and the tolerance conferred by ischemic preconditioning (IPC), would reveal new neuroprotective targets. We found that the product of the tuberous sclerosis complex 1 gene (TSC1), hamartin, is selectively induced by ischemia in hippocampal CA3 neurons. In CA1 neurons, hamartin was unaffected by ischemia but was upregulated by IPC preceding ischemia, which protects the otherwise vulnerable CA1 cells. Suppression of hamartin expression with TSC1 shRNA viral vectors both in vitro and in vivo increased the vulnerability of neurons to cell death following oxygen glucose deprivation (OGD) and ischemia. In vivo, suppression of TSC1 expression increased locomotor activity and decreased habituation in a hippocampal-dependent task. Overexpression of hamartin increased resistance to OGD by inducing productive autophagy through an mTORC1-dependent mechanism.


Subject(s)
Autophagy , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology , Hypoxia-Ischemia, Brain/prevention & control , Neuroprotective Agents/metabolism , Tumor Suppressor Proteins/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy/drug effects , CA1 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/metabolism , Cells, Cultured , Hypoxia , Hypoxia-Ischemia, Brain/metabolism , Ischemic Preconditioning , Male , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Prosencephalon/blood supply , Prosencephalon/injuries , Proteins/antagonists & inhibitors , Proteins/metabolism , RNA Interference , RNA, Small Interfering , Rats , Rats, Wistar , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/genetics
9.
PLoS One ; 8(1): e53472, 2013.
Article in English | MEDLINE | ID: mdl-23320088

ABSTRACT

Loss of integrity of the basal forebrain cholinergic neurons is a consistent feature of Alzheimer's disease, and measurement of basal forebrain degeneration by magnetic resonance imaging is emerging as a sensitive diagnostic marker for prodromal disease. It is also known that Alzheimer's disease patients perform poorly on both real space and computerized cued (allothetic) or uncued (idiothetic) recall navigation tasks. Although the hippocampus is required for allothetic navigation, lesions of this region only mildly affect idiothetic navigation. Here we tested the hypothesis that the cholinergic medial septo-hippocampal circuit is important for idiothetic navigation. Basal forebrain cholinergic neurons were selectively lesioned in mice using the toxin saporin conjugated to a basal forebrain cholinergic neuronal marker, the p75 neurotrophin receptor. Control animals were able to learn and remember spatial information when tested on a modified version of the passive place avoidance test where all extramaze cues were removed, and animals had to rely on idiothetic signals. However, the exploratory behaviour of mice with cholinergic basal forebrain lesions was highly disorganized during this test. By contrast, the lesioned animals performed no differently from controls in tasks involving contextual fear conditioning and spatial working memory (Y maze), and displayed no deficits in potentially confounding behaviours such as motor performance, anxiety, or disturbed sleep/wake cycles. These data suggest that the basal forebrain cholinergic system plays a specific role in idiothetic navigation, a modality that is impaired early in Alzheimer's disease.


Subject(s)
Cholinergic Neurons/physiology , Cues , Maze Learning/physiology , Prosencephalon/physiopathology , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Animals , Cholinergic Neurons/drug effects , Conditioning, Psychological/physiology , Fear/physiology , Humans , Locomotion/physiology , Male , Memory, Short-Term/physiology , Mental Recall/physiology , Mice , Mice, Inbred C57BL , Prosencephalon/drug effects , Prosencephalon/injuries , Ribosome Inactivating Proteins, Type 1/toxicity , Saporins
10.
J Neurosci ; 32(38): 13244-54, 2012 Sep 19.
Article in English | MEDLINE | ID: mdl-22993440

ABSTRACT

The basal forebrain (BF) is a key structure in regulating both cortical activity and sleep homeostasis. It receives input from all ascending arousal systems and is particularly highly innervated by histaminergic neurons. Previous studies clearly point to a role for histamine as a wake-promoting substance in the BF. We used in vivo microdialysis and pharmacological treatments in rats to study which electroencephalogram (EEG) spectral properties are associated with histamine-induced wakefulness and whether this wakefulness is followed by increased sleep and increased EEG delta power during sleep. We also investigated which BF neurons mediate histamine-induced cortical activation. Extracellular BF histamine levels rose immediately and remained constant throughout a 6 h period of sleep deprivation, returning to baseline levels immediately afterward. During the spontaneous sleep-wake cycle, we observed a strong correlation between wakefulness and extracellular histamine concentrations in the BF, which was unaffected by the time of day. The perfusion of histamine into the BF increased wakefulness and cortical activity without inducing recovery sleep. The perfusion of a histamine receptor 1 antagonist into the BF decreased both wakefulness and cortical activity. Lesioning the BF cholinergic neurons abolished these effects. Together, these results show that activation of the cholinergic BF by histamine is important in sustaining a high level of cortical activation, and that a lack of activation of the cholinergic BF by histamine may be important in initiating and maintaining nonrapid eye movement sleep. The level of histamine release is tightly connected to behavioral state, but conveys no information about sleep pressure.


Subject(s)
Cerebral Cortex/physiology , Cholinergic Neurons/physiology , Histamine Release/physiology , Prosencephalon/cytology , Prosencephalon/metabolism , Analysis of Variance , Animals , Antibodies, Monoclonal/toxicity , Cerebral Cortex/drug effects , Choline O-Acetyltransferase/metabolism , Cholinergic Agents/toxicity , Cholinergic Neurons/drug effects , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Electroencephalography , Electromyography , Fourier Analysis , Functional Laterality , Histamine/administration & dosage , Histamine Agonists/administration & dosage , Histamine Antagonists/pharmacology , Histamine Release/drug effects , Male , Microdialysis , Prosencephalon/drug effects , Prosencephalon/injuries , Rats , Rats, Wistar , Ribosome Inactivating Proteins, Type 1/toxicity , Saporins , Sleep Deprivation/physiopathology , Sleep Stages/drug effects , Sleep Stages/physiology , Time Factors , Wakefulness/drug effects
11.
J Neurosci ; 31(45): 16353-68, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22072687

ABSTRACT

Accurate timing is a critical aspect of motor control, yet the temporal structure of many mature behaviors emerges during learning from highly variable exploratory actions. How does a developing brain acquire the precise control of timing in behavioral sequences? To investigate the development of timing, we analyzed the songs of young juvenile zebra finches. These highly variable vocalizations, akin to human babbling, gradually develop into temporally stereotyped adult songs. We find that the durations of syllables and silences in juvenile singing are formed by a mixture of two distinct modes of timing: a random mode producing broadly distributed durations early in development, and a stereotyped mode underlying the gradual emergence of stereotyped durations. Using lesions, inactivations, and localized brain cooling, we investigated the roles of neural dynamics within two premotor cortical areas in the production of these temporal modes. We find that LMAN (lateral magnocellular nucleus of the nidopallium) is required specifically for the generation of the random mode of timing and that mild cooling of LMAN causes an increase in the durations produced by this mode. On the contrary, HVC (used as a proper name) is required specifically for producing the stereotyped mode of timing, and its cooling causes a slowing of all stereotyped components. These results show that two neural pathways contribute to the timing of juvenile songs and suggest an interesting organization in the forebrain, whereby different brain areas are specialized for the production of distinct forms of neural dynamics.


Subject(s)
Models, Neurological , Nerve Net/physiology , Neural Pathways/physiology , Nonlinear Dynamics , Prosencephalon/physiology , Vocalization, Animal , Animals , Behavior, Animal , Computer Simulation , Male , Nerve Net/injuries , Neural Pathways/injuries , Prosencephalon/anatomy & histology , Prosencephalon/injuries , Respiration , Songbirds , Sound Spectrography/methods , Spectrum Analysis , Stereotyped Behavior , Time Factors , Time Perception
12.
Brain Res ; 1373: 79-90, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21138733

ABSTRACT

Previous work in our laboratory indicated that cholinergic denervation by intraventricular infusion of 192-IgG-saporin on postnatal day 7 (N192S) reduced the number of cells in the dentate gyrus expressing doublecortin, a marker for immature neuroblasts. In addition, there was a suggestion that N192S impaired the neurogenic response to environmental enrichment (EE). The purpose of the present study was to further characterize the impact of N192S on the proliferation, differentiation and survival of newborn cells in the dentate gyrus. After 42 days in EE or standard housing, all rats received injections of 5-bromo-2-deoxyuridine (BrdU) to label dividing cells. They were sacrificed either one day (to assess cell proliferation) or 28 days later (to assess survival and differentiation of BrdU-labelled cells). EE failed to increase neurogenesis, thereby preventing determination of the effects of N192S on EE-induced neurogenesis. However, N192S by itself reduced the number of BrdU(+) cells 1 day after BrdU exposure, but did not alter the number of cells expressing the cell cycle marker Ki-67. The number of BrdU(+) cells 28 days after BrdU exposure was not affected by N192S. Confocal analysis of BrdU(+) cells double-immunofluorescently stained to detect NeuN or S100B indicated that N192S did not alter the proportion of new cells that adopted a neuronal or glial identity. The most plausible explanation for these results is that N192S accelerates the death of newborn cells, but does not change their overall survival rate or phenotypic differentiation.


Subject(s)
Antibodies, Monoclonal/toxicity , Cholinergic Agents/toxicity , Hippocampus/physiopathology , Neurogenesis/physiology , Prosencephalon/drug effects , Ribosome Inactivating Proteins, Type 1/toxicity , Acetylcholinesterase/metabolism , Age Factors , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Doublecortin Domain Proteins , Doublecortin Protein , Female , Ki-67 Antigen/metabolism , Male , Microtubule-Associated Proteins/metabolism , Nerve Growth Factors/metabolism , Neurogenesis/drug effects , Neuropeptides/metabolism , Prosencephalon/growth & development , Prosencephalon/injuries , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Saporins , Statistics as Topic
13.
Dev Neurosci ; 32(5-6): 374-84, 2010.
Article in English | MEDLINE | ID: mdl-20829579

ABSTRACT

In severe paediatric traumatic brain injury (TBI), a common focus of treatment is raised intracranial pressure (ICP). We have previously reported frontal cerebral vulnerability with executive deficits from raised ICP in paediatric TBI. Now, using diffusion tensor imaging (DTI) in a different population, we have examined fractional anisotropy (FA), and mean, axial and radial diffusivity (MD, AD, RD) in 4 regions of the corpus callosum (CC) and in both inferior frontal regions. Our aim was to examine during the chronic phase of TBI whether the CC cross-sectional area correlated with regional DTI metrics of white matter microstructure, with global outcome ratings of function (Functional Independence Measure and Multiattribute Health Status Classification) and with performance in the Rey-Osterrieth Complex Figure (ROCF) test. We examined 33 paediatric TBI cases who were followed, on average, 4.9 years after severe injury. All cases had received mechanical ventilation during their acute treatment and, a priori, they were assigned to a non-ICP or a raised ICP group. Twenty-two participants had mainly right-sided injury at the time of acute ictus. The findings confirm that severe TBI in childhood, complicated by intracranial hypertension, results in CC vulnerability. In the chronic phase of recovery, it is reduced in the cross-sectional area, it is more compact and thinned, and the anterior region is disproportionately small. Late after raised ICP, we have also found that individuals exhibit regional microstructural abnormality with combined reduced FA and increased MD, AD and RD. Smaller size and such microstructural changes in the anterior CC were associated with similar right-sided (rather than left-sided) frontal microstructural changes in the ICP group. Taken together, this evidence points to an interaction between raised ICP-related brain tissue perturbation and focal frontal extracallosal injury, leading to anterior CC regional vulnerability, most likely wallerian degeneration. At long-term follow-up, this lack of white matter integrity in the anterior CC is correlated with functional outcome, particularly in aspects of social interaction and the copy component of the ROCF test, which suggests that the CC-to-forebrain function warrants further study in chronic TBI.


Subject(s)
Brain Injuries/pathology , Corpus Callosum/pathology , Intracranial Hypertension/complications , Prosencephalon/pathology , Recovery of Function , Adolescent , Brain Injuries/physiopathology , Child , Corpus Callosum/injuries , Corpus Callosum/physiopathology , Diffusion Tensor Imaging , Humans , Image Interpretation, Computer-Assisted , Intracranial Hypertension/pathology , Intracranial Hypertension/physiopathology , Prosencephalon/injuries , Prosencephalon/physiopathology , Young Adult
14.
Microcirculation ; 17(6): 427-38, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20690981

ABSTRACT

EtOH-PC reduces postischemic neuronal injury in response to cerebral (I/R). We examined the mechanism underlying this protective effect by determining (i) whether it was associated with a decrease in I/R-induced leukocyte-endothelial adhesive interactions in postcapillary venules, and (ii) whether the protective effects were mediated by activation of large conductance, calcium-activated potassium (BK(Ca)) channels. Mice were administered ethanol by gavage or treated with the BK(Ca) channel opener, NS1619, 24 hours prior to I/R with or without prior treatment with the BK(Ca) channel blocker, PX. Both CCA were occluded for 20 minutes followed by two and three hours of reperfusion, and rolling (LR) and adherent (LA) leukocytes were quantified in pial venules using intravital microscopy. The extent of DND, apoptosis and glial activation in hippocampus were assessed four days after I/R. Compared with sham, I/R elicited increases in LR and LA in pial venules and DND and apoptosis as well as glial activation in the hippocampus. These effects were attenuated by EtOH-PC or antecedent NS1619 administration, and this protection was reversed by prior treatment with PX. Our results support a role for BK(Ca) channel activation in the neuroprotective effects of EtOH-PC in cerebral I/R.


Subject(s)
Brain Ischemia/drug therapy , Ethanol/administration & dosage , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Leukocytes/drug effects , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Reperfusion Injury/drug therapy , Animals , Benzimidazoles/pharmacology , Blood-Brain Barrier/drug effects , Brain Ischemia/blood , Brain Ischemia/pathology , Cell Adhesion/drug effects , Cell Death/drug effects , Cerebrovascular Circulation/drug effects , Endothelial Cells/drug effects , Indoles/pharmacology , Ischemic Preconditioning/methods , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Neurons/pathology , Prosencephalon/blood supply , Prosencephalon/drug effects , Prosencephalon/injuries , Reperfusion Injury/blood , Reperfusion Injury/pathology
15.
Gen Physiol Biophys ; 29(2): 113-21, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20577022

ABSTRACT

Although ischemia/reperfusion injury remains incompletely understood, it appears that reactive oxygen species produced mainly during postischemic recirculation play a critical role. The present study examined the impact of forebrain ischemia and subsequent one-day reperfusion on several blood parameters. We determined glutamate concentration in whole blood, measured Cu/Zn- and Mn-SOD (superoxide dismutase) activity in blood cells as well as plasma, and investigated the prevalence of single and double strand breaks of lymphocyte DNA. The results of our experiment showed that the concentration of glutamic acid in whole blood was increased by about 25%. Antioxidant activity of total SOD and Cu/Zn-SOD was reduced in blood cells and plasma. Mn-SOD activity in blood cells was not affected by ischemic insult and one-day reperfusion, but we detected its significantly lower activity in samples of plasma. We observed a weakly reduced level of double and a significantly elevated level of single strand breaks of lymphocyte DNA. In conclusion, one day of recovery after the ischemic attack failed to return peripheral circulatory system to physiological conditions. Reduced antioxidant capacity in the blood and an elevated level of excitotoxic amino acid glutamate may cause lymphocyte DNA damage, and probably contribute to insufficient postischemic recovery of brain tissue.


Subject(s)
Brain Ischemia/physiopathology , Prosencephalon/blood supply , Prosencephalon/injuries , Reperfusion Injury/physiopathology , Animals , Brain Ischemia/blood , Cerebrovascular Circulation/physiology , DNA Damage , Glutamic Acid/blood , Lymphocytes/metabolism , Male , Rats , Rats, Wistar , Reperfusion Injury/blood , Superoxide Dismutase/blood
16.
J Neurosci ; 30(21): 7392-400, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20505106

ABSTRACT

Motor exploration can be an adaptive strategy when behavior fails to achieve an expected outcome. For example, like humans, adult songbirds change their vocal output when auditory feedback is altered or absent. Here, we show that the output of an anterior forebrain pathway (AFP) through the avian basal ganglia directly contributes to the expression of deafening-induced vocal changes in adulthood. Lesioning the output nucleus of this circuit in adult male zebra finches reverses moderate changes in song structure and variability caused by deafening. Furthermore, the results indicate that more severe deafening-induced changes in vocal behavior likely reflect altered function outside the AFP (e.g., within the vocal motor pathway). AFP lesions do not promote recovery if songs are severely deteriorated at the time of lesion even though previous work shows that the AFP is required for such deterioration to emerge. Thus, in birds, as in mammals, the contribution of basal ganglia-thalamic-cortical circuits to motor control may change when feedback is absent or unexpected and includes both "active" and "permissive" roles.


Subject(s)
Auditory Pathways/physiology , Basal Ganglia/physiology , Deafness/physiopathology , Prosencephalon/physiology , Vocalization, Animal/physiology , Analysis of Variance , Animals , Auditory Pathways/injuries , Behavior, Animal , Feedback, Sensory , Finches/physiology , Learning/physiology , Male , Models, Biological , Prosencephalon/injuries , Sound Spectrography/methods , Time Factors
17.
J Neurophysiol ; 102(2): 979-91, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19515953

ABSTRACT

In adult male zebra finches, transecting the vocal nerve causes previously stable (i.e., crystallized) song to slowly degrade, presumably because of the resulting distortion in auditory feedback. How and where distorted feedback interacts with song motor networks to induce this process of song decrystallization remains unknown. The song premotor nucleus HVC is a potential site where auditory feedback signals could interact with song motor commands. Although the forebrain nucleus interface of the nidopallium (NIf) appears to be the primary auditory input to HVC, NIf lesions made in adult zebra finches do not trigger song decrystallization. One possibility is that NIf lesions do not interfere with song maintenance, but do compromise the adult zebra finch's ability to express renewed vocal plasticity in response to feedback perturbations. To test this idea, we bilaterally lesioned NIf and then transected the vocal nerve in adult male zebra finches. We found that bilateral NIf lesions did not prevent nerve section-induced song decrystallization. To test the extent to which the NIf lesions disrupted auditory processing in the song system, we made in vivo extracellular recordings in HVC and a downstream anterior forebrain pathway (AFP) in NIf-lesioned birds. We found strong and selective auditory responses to the playback of the birds' own song persisted in HVC and the AFP following NIf lesions. These findings suggest that auditory inputs to the song system other than NIf, such as the caudal mesopallium, could act as a source of auditory feedback signals to the song motor network.


Subject(s)
Brain/physiology , Finches/physiology , Vocalization, Animal/physiology , Action Potentials , Animals , Auditory Pathways/physiology , Auditory Perception/physiology , Feedback, Psychological/physiology , Functional Laterality , Male , Microelectrodes , Neural Pathways/injuries , Neural Pathways/physiology , Prosencephalon/injuries , Prosencephalon/physiology , Sound Spectrography , Time Factors
18.
J Neurophysiol ; 101(6): 3235-45, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19357331

ABSTRACT

Behavioral variability is important for motor skill learning but continues to be present and actively regulated even in well-learned behaviors. In adult songbirds, two types of song variability can persist and are modulated by social context: variability in syllable structure and variability in syllable sequencing. The degree to which the control of both types of adult variability is shared or distinct remains unknown. The output of a basal ganglia-forebrain circuit, LMAN (the lateral magnocellular nucleus of the anterior nidopallium), has been implicated in song variability. For example, in adult zebra finches, neurons in LMAN actively control the variability of syllable structure. It is unclear, however, whether LMAN contributes to variability in adult syllable sequencing because sequence variability in adult zebra finch song is minimal. In contrast, Bengalese finches retain variability in both syllable structure and syllable sequencing into adulthood. We analyzed the effects of LMAN lesions on the variability of syllable structure and sequencing and on the social modulation of these forms of variability in adult Bengalese finches. We found that lesions of LMAN significantly reduced the variability of syllable structure but not of syllable sequencing. We also found that LMAN lesions eliminated the social modulation of the variability of syllable structure but did not detect significant effects on the modulation of sequence variability. These results show that LMAN contributes differentially to syllable versus sequence variability of adult song and suggest that these forms of variability are regulated by distinct neural pathways.


Subject(s)
Basal Ganglia/physiology , Finches/physiology , Prosencephalon/physiology , Sexual Behavior, Animal/physiology , Social Behavior , Vocalization, Animal/physiology , Animal Communication , Animals , Basal Ganglia/injuries , Behavior, Animal , Male , Nerve Net/physiology , Neural Pathways/injuries , Neural Pathways/physiology , Prosencephalon/injuries
19.
Mol Cells ; 27(4): 397-401, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19390819

ABSTRACT

Olig2 transcription factor is widely expressed throughout the central nervous system; therefore, it is considered to have multiple functions in the developing, mature and injured brain. In this mini-review, we focus on Olig2 in the forebrain (telencephalon and diencephalon) and discuss the functional significance of Olig2 and the differentiation properties of Olig2-expressing progenitors in the development and injured states. Short- and long-term lineage analysis in the developing forebrain elucidated that not all late Olig2+ cells are direct cohorts of early cells and that Olig2 lineage cells differentiate into neurons or glial cells in a region- and stage-dependent manner. Olig2-deficient mice revealed large elimination of oligodendrocyte precursor cells and a decreased number of astrocyte progenitors in the dorsal cortex, whereas no reduction in the number of GABAergic neurons. In addition to Olig2 function in the developing cortex, Olig2 is also reported to be important for glial scar formation after injury. Thus, Olig2 can be essential for glial differentiation during development and after injury.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Nerve Tissue Proteins/physiology , Neuroglia/physiology , Prosencephalon/physiology , Animals , Cell Lineage , Humans , Mice , Oligodendrocyte Transcription Factor 2 , Prosencephalon/cytology , Prosencephalon/growth & development , Prosencephalon/injuries , Stem Cells/physiology
20.
Brain Res ; 1252: 172-82, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19084506

ABSTRACT

Intraventricular injections of 192 IgG saporin in 7-day-old rat severely reduced hippocampal cholinergic innervation as reflected by both decreased acetylcholinesterase staining and immunoreactivity for the p75 neurotrophin receptor. It was determined if this altered the effects of environmental enrichment on spatial learning, hippocampal CA1 cell cytoarchitecture as reflected by the Golgi stain, and neurogenesis in the dentate gyrus as indicated by doublecortin immunoreactivity. At weaning, lesioned and control rats were either group housed in large, environmentally enriched cages or housed two per standard cage for 42 days. When subsequently assessed with a working-memory spatial navigation task, both lesioned and control rats showed enhanced learning as a result of enrichment. Quantitative analysis of Golgi stained sections indicated that enrichment did not affect CA1 dendritic branching, total dendritic length or dendritic spine density. However, the lesion reduced the number of apical branches, spine density on intermediate to distal apical dendrites, and the length of basal branches. It also reduced the number of doublecortin immunoreactive neurons in the dentate gyrus and appeared to prevent their increase due to environmental enrichment. It is concluded that developmental cholinergic lesioning does not attenuate neurobehavioral plasticity, at least as reflected by the behavioral consequences of enrichment. It does, however, attenuate neurogenesis in the dentate gyrus, like adult-inflicted cholinergic lesions. As previously found for cortical neurons, it also reduces CA1 pyramidal cell dendritic complexity and spine density in adulthood. The results have implications for the loss of synapses that occurs in both developmental and aging-related brain disorders involving cholinergic dysfunction.


Subject(s)
Acetylcholine/metabolism , Brain Injuries/physiopathology , Environment , Hippocampus/cytology , Neurogenesis , Prosencephalon/injuries , Prosencephalon/physiopathology , Analysis of Variance , Animals , Antibodies, Monoclonal/pharmacology , Behavior, Animal/physiology , Brain Injuries/chemically induced , Brain Injuries/therapy , Dendritic Spines , Doublecortin Domain Proteins , Doublecortin Protein , Female , Hippocampus/physiopathology , Immunohistochemistry , Male , Maze Learning , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Neurotoxins/pharmacology , Prosencephalon/anatomy & histology , Pyramidal Cells/physiopathology , Pyramidal Cells/ultrastructure , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins
SELECTION OF CITATIONS
SEARCH DETAIL
...