Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Rep ; 26(9): 2477-2493.e9, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30811995

ABSTRACT

The role of brain cell-type-specific functions and profiles in pathological and non-pathological contexts is still poorly defined. Such cell-type-specific gene expression profiles in solid, adult tissues would benefit from approaches that avoid cellular stress during isolation. Here, we developed such an approach and identified highly selective transcriptomic signatures in adult mouse striatal direct and indirect spiny projection neurons, astrocytes, and microglia. Integrating transcriptomic and epigenetic data, we obtained a comprehensive model for cell-type-specific regulation of gene expression in the mouse striatum. A cross-analysis with transcriptomic and epigenomic data generated from mouse and human Huntington's disease (HD) brains shows that opposite epigenetic mechanisms govern the transcriptional regulation of striatal neurons and glial cells and may contribute to pathogenic and compensatory mechanisms. Overall, these data validate this less stressful method for the investigation of cellular specificity in the adult mouse brain and demonstrate the potential of integrative studies using multiple databases.


Subject(s)
Brain/metabolism , Huntington Disease/genetics , Animals , DNA/chemistry , Epigenesis, Genetic , Gene Expression Profiling/methods , Humans , Huntington Disease/metabolism , Laser Capture Microdissection/methods , Male , Mice , Mice, Transgenic , MicroRNAs/metabolism , Nucleic Acid Conformation , RNA, Messenger/metabolism , Transcription Factors/metabolism
2.
Neurobiol Dis ; 86: 131-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26626081

ABSTRACT

Huntington's disease (HD) is an autosomal dominant disease that develops in midlife (~ 40 years-old at onset) and then progresses slowly. It is still unclear how striatal medium spiny neurons (MSNs), the most vulnerable neurons in HD, maintain their function for decades despite the chronic expression of mutant huntingtin (mHTT). In this study, we used aged BACHD mice, a HD model expressing the full-length human mHTT gene, to investigate the molecular, morphological and functional properties of striatal MSNs. We report that the density of dendritic spines in MSNs is substantially lower in aged BACHD mice than in wild-type (WT) mice, in the absence of major dendritic changes and neuronal loss. This spine loss is accompanied by changes in transcription, resulting in a low expression of the striatum-specific G protein-coupled receptor 88 (Gpr88) as well as a reorganization of the composition of AMPAR subunits (high Gria1/Gria2 mRNA ratio). We also detected functional changes in BACHD MSNs. Notably, BACHD MSNs were hyperexcitable and the amplitude of AMPAR-mediated synaptic currents was higher than in WT MSNs. Altogether, these data show that both the intrinsic properties and the strength of the remaining synapses are modified in MSNs with low dendritic spine density in aged BACHD mice. These homeostatic mechanisms may compensate for the substantial loss of synaptic inputs and thus alleviate the deleterious effects of mHTT expression on the activity of MSNs and also possibly on the motor phenotype in aged BACHD.


Subject(s)
Corpus Striatum/pathology , Corpus Striatum/physiopathology , Huntington Disease/pathology , Huntington Disease/physiopathology , Neurons/pathology , Neurons/physiology , Synapses/physiology , Animals , Corpus Striatum/metabolism , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disease Models, Animal , Disease Progression , Excitatory Postsynaptic Potentials , Female , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/metabolism , Protein Subunits/metabolism , Receptors, AMPA/metabolism , Receptors, G-Protein-Coupled/metabolism , Synapses/metabolism
3.
Acta Neuropathol ; 124(6): 777-95, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22976049

ABSTRACT

Whole-cell patch-clamp recordings and high-resolution morphometry were used to assess functional and structural properties of layer 3 pyramidal neurons in early (<4 months) and advanced (>8 months) stages of tauopathy in frontal cortical slices prepared from rTg4510 tau mutant (P301L) mice. In early tauopathy, dendritic architecture is preserved. In advanced tauopathy, neurons can be categorized as either "atrophic" (58 %)-exhibiting marked atrophy of the apical tuft, or "intact" (42 %)-with normal apical tufts and, in some instances, proliferative sprouting of oblique branches of the apical trunk. Approximately equal numbers of atrophic and intact neurons contain neurofibrillary tangles (NFTs) or are tangle-free, lending further support to the idea that NFTs per se are not toxic. Spine density is decreased due to a specific reduction in mushroom spines, but filopodia are increased in both atrophic and intact neurons. By contrast to these morphological changes, which are robust only in the advanced stage, significant electrophysiological changes are present in the early stage and persist in the advanced stage in both atrophic and intact neurons. The most marked of these changes are: a depolarized resting membrane potential, an increased depolarizing sag potential and increased action potential firing rates-all indicative of hyperexcitability. Spontaneous excitatory postsynaptic currents are not reduced in frequency or amplitude in either stage. The difference in the time course of functionally important electrophysiological changes versus regressive morphological changes implies differences in pathogenic mechanisms underlying functional and structural changes to neurons during progressive tauopathy.


Subject(s)
Electrophysiological Phenomena/physiology , Frontal Lobe/pathology , Membrane Potentials/physiology , Pyramidal Cells/pathology , Tauopathies/physiopathology , Action Potentials/physiology , Animals , Dendrites/pathology , Dendrites/physiology , Disease Models, Animal , Excitatory Postsynaptic Potentials/physiology , Mice , Mice, 129 Strain , Mice, Transgenic , Patch-Clamp Techniques/methods , Pyramidal Cells/physiology , Tauopathies/pathology
4.
Acta Neuropathol ; 122(5): 551-64, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21968531

ABSTRACT

Cortical neuron death is prevalent by 9 months in rTg(tau(P301L))4510 tau mutant mice (TG) and surviving pyramidal cells exhibit dendritic regression and spine loss. We used whole-cell patch-clamp recordings to investigate the impact of these marked structural changes on spontaneous excitatory and inhibitory postsynaptic currents (sEPSCs and sIPSCs) of layer 3 pyramidal cells in frontal cortical slices from behaviorally characterized TG and non-transgenic (NT) mice at this age. Frontal lobe function of TG mice was intact following a short delay interval but impaired following a long delay interval in an object recognition test, and cortical atrophy and cell loss were pronounced. Surviving TG cells had significantly reduced dendritic diameters, total spine density, and mushroom spines, yet sEPSCs were increased and sIPSCs were unchanged in frequency. Thus, despite significant regressive structural changes, synaptic responses were not reduced in TG cells, indicating that homeostatic compensatory mechanisms occur during progressive tauopathy. Consistent with this idea, surviving TG cells were more intrinsically excitable than NT cells, and exhibited sprouting of filopodia and axonal boutons. Moreover, the neuropil in TG mice showed an increased density of asymmetric synapses, although their mean size was reduced. Taken together, these data indicate that during progressive tauopathy, cortical pyramidal cells compensate for loss of afferent input by increased excitability and establishment of new synapses. These compensatory homeostatic mechanisms may play an important role in slowing the progression of neuronal network dysfunction during neurodegenerative tauopathies.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Homeostasis/physiology , Inhibitory Postsynaptic Potentials/physiology , Pyramidal Cells/physiopathology , Tauopathies/physiopathology , Animals , Cognition/physiology , Disease Models, Animal , Disease Progression , Frontal Lobe/metabolism , Frontal Lobe/pathology , Mice , Mice, Mutant Strains , Patch-Clamp Techniques , Pyramidal Cells/pathology , Synapses/physiology , Tauopathies/pathology , tau Proteins/genetics , tau Proteins/metabolism
5.
Brain Struct Funct ; 214(2-3): 181-99, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20177698

ABSTRACT

In neurodegenerative disorders, such as Alzheimer's disease, neuronal dendrites and dendritic spines undergo significant pathological changes. Because of the determinant role of these highly dynamic structures in signaling by individual neurons and ultimately in the functionality of neuronal networks that mediate cognitive functions, a detailed understanding of these changes is of paramount importance. Mutant murine models, such as the Tg2576 APP mutant mouse and the rTg4510 tau mutant mouse have been developed to provide insight into pathogenesis involving the abnormal production and aggregation of amyloid and tau proteins, because of the key role that these proteins play in neurodegenerative disease. This review showcases the multidimensional approach taken by our collaborative group to increase understanding of pathological mechanisms in neurodegenerative disease using these mouse models. This approach includes analyses of empirical 3D morphological and electrophysiological data acquired from frontal cortical pyramidal neurons using confocal laser scanning microscopy and whole-cell patch-clamp recording techniques, combined with computational modeling methodologies. These collaborative studies are designed to shed insight on the repercussions of dystrophic changes in neocortical neurons, define the cellular phenotype of differential neuronal vulnerability in relevant models of neurodegenerative disease, and provide a basis upon which to develop meaningful therapeutic strategies aimed at preventing, reversing, or compensating for neurodegenerative changes in dementia.


Subject(s)
Alzheimer Disease/pathology , Cerebral Cortex/pathology , Dendrites/pathology , Electrophysiology/methods , Image Processing, Computer-Assisted/methods , Pyramidal Cells/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Computer Simulation , Dendrites/metabolism , Disease Models, Animal , Mice , Microscopy, Confocal/methods , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Patch-Clamp Techniques/methods , Pyramidal Cells/metabolism , Pyramidal Cells/physiopathology , Staining and Labeling/methods
6.
Am J Pathol ; 176(1): 353-68, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20008141

ABSTRACT

Mutations in the presenilin 1 (PS1) gene are the most commonly recognized cause of familial Alzheimer's disease (FAD). Besides senile plaques, neurofibrillary tangles, and neuronal loss, Alzheimer's disease (AD) is also accompanied by vascular pathology. Here we describe an age-related vascular pathology in two lines of PS1 FAD-mutant transgenic mice that mimics many features of the vascular pathology seen in AD. The pathology was especially prominent in the microvasculature whose vessels became thinned and irregular with the appearance of many abnormally looped vessels as well as string vessels. Stereologic assessments revealed a reduction of the microvasculature in the hippocampus that was accompanied by hippocampal atrophy. The vascular changes were not congophilic. Yet, despite the lack of congophilia, penetrating vessels at the cortical surface were often abnormal morphologically and microhemorrhages sometimes occurred. Altered immunostaining of blood vessels with basement membrane-associated antigens was an early feature of the microangiopathy and was associated with thickening of the vascular basal laminae and endothelial cell alterations that were visible ultrastructurally. Interestingly, although the FAD-mutant transgene was expressed in neurons in both lines of mice, there was no detectable expression in vascular endothelial cells or glial cells. These studies thus have implications for the role of neuronal to vascular signaling in the pathogenesis of the vascular pathology associated with AD.


Subject(s)
Aging/pathology , Alzheimer Disease/genetics , Blood Vessels/pathology , Mutation/genetics , Presenilin-1/metabolism , Aging/metabolism , Animals , Atrophy , Basement Membrane/metabolism , Blood Vessels/abnormalities , Blood Vessels/metabolism , Blood Vessels/ultrastructure , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Chromosomes, Artificial, P1 Bacteriophage/genetics , Dendrites/metabolism , Dendrites/pathology , Extracellular Matrix Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/abnormalities , Microvessels/metabolism , Microvessels/pathology , Microvessels/ultrastructure , Mutant Proteins/metabolism , Transgenes/genetics
7.
Neurobiol Aging ; 30(5): 782-92, 2009 May.
Article in English | MEDLINE | ID: mdl-17919783

ABSTRACT

Aging is associated with deficiencies in the prefrontal cortex, including working memory impairment and compromised integrity of neuronal dendrites. Although protein kinase C (PKC) is implicated in structural plasticity, and overactivation of PKC results in working memory impairments in young animals, the role of PKC in prefrontal cortical impairments in the aged has not been examined. This study provides the first evidence that PKC activity is associated with prefrontal cortical dysfunction in aging. Pharmacological inhibition of PKC with chelerythrine rescued working memory impairments in aged rats and enhanced working memory in aged rhesus monkeys. Improvement correlated with age, with older monkeys demonstrating a greater degree of improvement following PKC inhibition. Furthermore, PKC activity within the prefrontal cortex was inversely correlated with the length of basal dendrites of prefrontal cortical neurons, as well as with working memory performance in aged rats. Together these findings indicate that PKC is dysregulated in aged animals and that PKC inhibitors may be useful in the treatment of cognitive deficits in the elderly.


Subject(s)
Aging/metabolism , Atrophy/enzymology , Cognition Disorders/enzymology , Prefrontal Cortex/enzymology , Protein Kinase C/metabolism , Aging/pathology , Aging/psychology , Animals , Atrophy/pathology , Atrophy/physiopathology , Benzophenanthridines/pharmacology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Dendrites/enzymology , Dendrites/pathology , Dendritic Spines/enzymology , Dendritic Spines/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Macaca mulatta , Male , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Neurons/enzymology , Neurons/pathology , Neuropsychological Tests , Prefrontal Cortex/pathology , Prefrontal Cortex/physiopathology , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
8.
Neurobiol Dis ; 32(2): 309-18, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18721884

ABSTRACT

Amyloid-beta (Abeta) plays a key role in the etiology of Alzheimer's disease, and pyramidal cell dendrites exposed to Abeta exhibit dramatic structural alterations, including reduced dendritic spine densities. To determine whether such structural alterations lead to electrophysiological changes, whole-cell patch clamp recordings with biocytin filling were used to assess both the electrophysiological and morphological properties of layer 3 pyramidal cells in frontal cortical slices prepared from 12-month-old Tg2576 amyloid precursor protein (APP) mutant vs. wild-type (Wt) mice. Tg2576 cells exhibited significantly increased dendritic lengths and volumes and decreased spine densities, while the total number of spines was not different from Wt. Tg2576 and Wt cells did not differ with regard to passive membrane, action potential firing or glutamatergic spontaneous excitatory postsynaptic current properties. Thus, overexpression of mutated APP in young Tg2576 mice leads to significant changes in neuronal morphological properties which do not have readily apparent functional consequences.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cerebral Cortex/pathology , Neurons/pathology , Neurons/physiology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Cell Size , Dendrites/pathology , Dendrites/physiology , Dendrites/ultrastructure , Disease Models, Animal , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Humans , In Vitro Techniques , Membrane Potentials/genetics , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neurons/ultrastructure , Patch-Clamp Techniques , Streptavidin/metabolism
9.
Genesis ; 46(6): 308-17, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18543298

ABSTRACT

The Cre-loxP system is widely used for making conditional alterations to the mouse genome. Cre-mediated recombination is frequently monitored using reporter lines in which Cre expression activates a reporter gene driven by a ubiquitous promoter. Given the distinct advantages of fluorescent reporters, we developed a transgenic reporter line, termed IRG, in which DsRed-Express, a red fluorescent protein (RFP) is expressed ubiquitously prior to Cre-mediated recombination and an enhanced green fluorescent protein (EGFP) following recombination. Besides their utility for monitoring Cre-mediated recombination, we show that in IRG mice red and green native fluorescence can be imaged simultaneously in thick tissue sections by confocal microscopy allowing for complex reconstructions to be created that are suitable for analysis of neuronal morphologies as well as neurovascular interactions in brain. IRG mice should provide a versatile tool for analyzing complex cellular relationships in both neural and nonneural tissues.


Subject(s)
Fluorescent Dyes/metabolism , Green Fluorescent Proteins/genetics , Integrases/metabolism , Luminescent Proteins/genetics , Recombination, Genetic , Animals , Brain/cytology , Brain/metabolism , Cells, Cultured , Crosses, Genetic , Diagnostic Imaging , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Genes, Reporter , Genetic Markers/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Integrases/genetics , Intermediate Filament Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Nerve Tissue Proteins/genetics , Nestin , Promoter Regions, Genetic , Tissue Distribution , Transgenes , Red Fluorescent Protein
10.
J Comp Neurol ; 507(1): 1141-50, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18157834

ABSTRACT

Anatomical alterations in the medial prefrontal cortex (mPFC) are associated with hypothalamopituitary adrenal (HPA) axis dysregulation, altered stress hormone levels, and psychiatric symptoms of stress-related mental illnesses. Functional imaging studies reveal impairment and shrinkage of the mPFC in such conditions, and these findings are paralleled by experimental studies showing dendritic retraction and spine loss following repeated stress in rodents. Here we extend this characterization to how repeated stress affects dendritic spine morphology in mPFC through the utilization of an automated approach that rapidly digitizes, reconstructs three dimensionally, and calculates geometric features of neurons. Rats were perfused after being subjected to 3 weeks of daily restraint stress (6 hours/day), and intracellular injections of Lucifer Yellow were made in layer II/III pyramidal neurons in the dorsal mPFC. To reveal spines in all angles of orientation, deconvolved high-resolution confocal laser scanning microscopy image stacks of dendritic segments were reconstructed and analyzed for spine volume, surface area, and length using a Rayburst-based automated approach (8,091 and 8,987 spines for control and stress, respectively). We found that repeated stress results in an overall decrease in mean dendritic spine volume and surface area, which was most pronounced in the distal portion of apical dendritic fields. Moreover, we observed an overall shift in the population of spines, manifested by a reduction in large spines and an increase in small spines. These results suggest a failure of spines to mature and stabilize following repeated stress and are likely to have major repercussions on function, receptor expression, and synaptic efficacy.


Subject(s)
Dendritic Spines/physiology , Neuronal Plasticity/physiology , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Stress, Physiological/physiopathology , Animals , Cell Shape/physiology , Male , Pyramidal Cells/physiology , Pyramidal Cells/ultrastructure , Rats , Rats, Sprague-Dawley
11.
Neurobiol Aging ; 29(9): 1296-307, 2008 Sep.
Article in English | MEDLINE | ID: mdl-17420070

ABSTRACT

The loss of presynaptic markers is thought to represent a strong pathologic correlate of cognitive decline in Alzheimer's disease (AD). Spinophilin is a postsynaptic marker mainly located to the heads of dendritic spines. We assessed total numbers of spinophilin-immunoreactive puncta in the CA1 and CA3 fields of hippocampus and area 9 in 18 elderly individuals with various degrees of cognitive decline. The decrease in spinophilin-immunoreactivity was significantly related to both Braak neurofibrillary tangle (NFT) staging and clinical severity but not A beta deposition staging. The total number of spinophilin-immunoreactive puncta in CA1 field and area 9 were significantly related to MMSE scores and predicted 23.5 and 61.9% of its variability. The relationship between total number of spinophilin-immunoreactive puncta in CA1 field and MMSE scores did not persist when adjusting for Braak NFT staging. In contrast, the total number of spinophilin-immunoreactive puncta in area 9 was still significantly related to the cognitive outcome explaining an extra 9.6% of MMSE and 25.6% of the Clinical Dementia Rating scores variability. Our data suggest that neocortical dendritic spine loss is an independent parameter to consider in AD clinicopathologic correlations.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Dendritic Spines/metabolism , Dendritic Spines/pathology , Hippocampus/metabolism , Hippocampus/pathology , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Aged , Biomarkers/metabolism , Disease Progression , Female , Humans , Immunohistochemistry , Male , Nerve Net/metabolism , Nerve Net/pathology , Tissue Distribution
12.
Cell Res ; 17(7): 619-26, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17593907

ABSTRACT

Neurovascular interactions are crucial for the normal development of the central nervous system. To study such interactions in primary cultures, we developed a procedure to simultaneously isolate neural progenitor and endothelial cell fractions from embryonic mouse brains. Depending on the culture conditions endothelial cells were found to favor maintenance of the neuroprogenitor phenotype through the production of soluble factors, or to promote neuronal differentiation of neural progenitors through direct contact. These apparently opposing effects could reflect differential cellular interactions needed for the proper development of the brain.


Subject(s)
Brain/cytology , Cell Differentiation/physiology , Endothelial Cells/cytology , Neuroepithelial Cells/cytology , Stem Cells/cytology , Animals , Brain/embryology , Brain/metabolism , Cells, Cultured , Coculture Techniques , Endothelial Cells/metabolism , Fibronectins/metabolism , Mice , Microscopy, Confocal , Neuroepithelial Cells/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Stem Cells/metabolism , von Willebrand Factor/metabolism
13.
Aging Cell ; 6(3): 275-84, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17465981

ABSTRACT

Structural changes of neurons in the brain during aging are complex and not well understood. Neurons have significant homeostatic control of essential brain functions, including synaptic excitability, gene expression, and metabolic regulation. Any deviations from the norm can have severe consequences as seen in aging and injury. In this review, we present some of the structural adaptations that neurons undergo throughout normal and pathological aging and discuss their effects on electrophysiological properties and cognition. During aging, it is evident that neurons undergo morphological changes such as a reduction in the complexity of dendrite arborization and dendritic length. Spine numbers are also decreased, and because spines are the major sites for excitatory synapses, changes in their numbers could reflect a change in synaptic densities. This idea has been supported by studies that demonstrate a decrease in the overall frequency of spontaneous glutamate receptor-mediated excitatory responses, as well as a decrease in the levels of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid and N-methyl-d-aspartate receptor expression. Other properties such as gamma-aminobutyric acid A receptor-mediated inhibitory responses and action potential firing rates are both significantly increased with age. These findings suggest that age-related neuronal dysfunction, which must underlie observed decline in cognitive function, probably involves a host of other subtle changes within the cortex that could include alterations in receptors, loss of dendrites, and spines and myelin dystrophy, as well as the alterations in synaptic transmission. Together these multiple alterations in the brain may constitute the substrate for age-related loss of cognitive function.


Subject(s)
Aging , Brain/metabolism , Brain/pathology , Action Potentials , Aged , Alzheimer Disease/metabolism , Dendrites/metabolism , Dendritic Spines/metabolism , Electrophysiology , Homeostasis , Humans , Models, Biological , Models, Neurological , Neurons/metabolism , Receptors, Glutamate/metabolism , gamma-Aminobutyric Acid/metabolism
14.
J Neurosci Methods ; 163(1): 76-82, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17403541

ABSTRACT

While the brain vasculature can be imaged with many methods, immunohistochemistry has distinct advantages due to its simplicity and applicability to archival tissue. However, immunohistochemical staining of the murine brain vasculature in aldehyde fixed tissue has proven elusive and inconsistent using current protocols. Here we investigated whether antigen retrieval methods could improve vascular staining in the adult mouse brain. We found that pepsin digestion prior to immunostaining unmasked widespread collagen IV staining of the cerebrovasculature in the adult mouse brain. Pepsin treatment also unmasked widespread vascular staining with laminin, but only marginally improved isolectin B4 staining and did not enhance vascular staining with fibronectin, perlecan or CD146. Collagen IV immunoperoxidase staining was easily combined with cresyl violet counterstaining making it suitable for stereological analyses of both vascular and neuronal parameters in the same tissue section. This method should be widely applicable for labeling the brain vasculature of the mouse in aldehyde fixed tissue from both normal and pathological states.


Subject(s)
Blood Vessels/drug effects , Blood Vessels/metabolism , Brain/anatomy & histology , Collagen Type IV/metabolism , Gastrointestinal Agents/pharmacology , Pepsin A/pharmacology , Animals , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry/methods , Indoles , Laminin/metabolism , Lectins/metabolism , Male , Mice , Mice, Inbred C57BL
15.
J Neurosci ; 26(30): 7870-4, 2006 Jul 26.
Article in English | MEDLINE | ID: mdl-16870732

ABSTRACT

Stressful life events have been implicated clinically in the pathogenesis of mental illness, but the neural substrates that may account for this observation remain poorly understood. Attentional impairments symptomatic of these psychiatric conditions are associated with structural and functional abnormalities in a network of prefrontal cortical structures. Here, we examine whether chronic stress-induced dendritic alterations in the medial prefrontal cortex (mPFC) and orbital frontal cortex (OFC) underlie impairments in the behaviors that they subserve. After 21 d of repeated restraint stress, rats were tested on a perceptual attentional set-shifting task, which yields dissociable measures of reversal learning and attentional set-shifting, functions that are mediated by the OFC and mPFC, respectively. Intracellular iontophoretic injections of Lucifer yellow were performed in a subset of these rats to examine dendritic morphology in layer II/III pyramidal cells of the mPFC and lateral OFC. Chronic stress induced a selective impairment in attentional set-shifting and a corresponding retraction (20%) of apical dendritic arbors in the mPFC. In stressed rats, but not in controls, decreased dendritic arborization in the mPFC predicted impaired attentional set-shifting performance. In contrast, stress was not found to adversely affect reversal learning or dendritic morphology in the lateral OFC. Instead, apical dendritic arborization in the OFC was increased by 43%. This study provides the first direct evidence that dendritic remodeling in the prefrontal cortex may underlie the functional deficits in attentional control that are symptomatic of stress-related mental illnesses.


Subject(s)
Attention Deficit Disorder with Hyperactivity/pathology , Dendrites/pathology , Perceptual Disorders/pathology , Prefrontal Cortex/pathology , Stress, Psychological/pathology , Animals , Attention Deficit Disorder with Hyperactivity/physiopathology , Male , Memory, Short-Term , Perceptual Disorders/physiopathology , Prefrontal Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Stress, Psychological/physiopathology
16.
Cereb Cortex ; 16(3): 313-20, 2006 Mar.
Article in English | MEDLINE | ID: mdl-15901656

ABSTRACT

The prefrontal cortex (PFC) plays an important role in higher cognitive processes, and in the regulation of stress-induced hypothalamic-pituitary-adrenal (HPA) activity. Here we examined the effect of repeated restraint stress on dendritic spine number in the medial PFC. Rats were perfused after receiving 21 days of daily restraint stress, and intracellular iontophoretic injections of Lucifer Yellow were carried out in layer II/III pyramidal neurons in the anterior cingulate and prelimbic cortices. We found that stress results in a significant (16%) decrease in apical dendritic spine density in medial PFC pyramidal neurons, and confirmed a previous observation that total apical dendritic length is reduced by 20% in the same neurons. We estimate that nearly one-third of all axospinous synapses on apical dendrites of pyramidal neurons in medial PFC are lost following repeated stress. A decrease in medial PFC dendritic spines may not only be indicative of a decrease in the total population of axospinous synapses, but may impair these neurons' capacity for biochemical compartmentalization and plasticity in which dendritic spines play a major role. Dendritic atrophy and spine loss may be important cellular features of stress-related psychiatric disorders where the PFC is functionally impaired.


Subject(s)
Dendrites/pathology , Prefrontal Cortex/pathology , Stress, Physiological/pathology , Animals , Male , Prefrontal Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Recurrence , Restraint, Physical/adverse effects , Stress, Physiological/etiology , Stress, Physiological/physiopathology
17.
Development ; 132(17): 3873-83, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16079160

ABSTRACT

Mice with a null mutation of the presenilin 1 gene (Psen1(-/-)) die during late intrauterine life or shortly after birth and exhibit multiple CNS and non-CNS abnormalities, including cerebral hemorrhages and altered cortical development. The cellular and molecular basis for the developmental effects of Psen1 remain incompletely understood. Psen1 is expressed in neural progenitors in developing brain, as well as in postmitotic neurons. We crossed transgenic mice with either neuron-specific or neural progenitor-specific expression of Psen1 onto the Psen1(-/-) background. We show that neither neuron-specific nor neural progenitor-specific expression of Psen1 can rescue the embryonic lethality of the Psen1(-/-) embryo. Indeed neuron-specific expression rescued none of the abnormalities in Psen1(-/-) mice. However, Psen1 expression in neural progenitors rescued the cortical lamination defects, as well as the cerebral hemorrhages, and restored a normal vascular pattern in Psen1(-/-) embryos. Collectively, these studies demonstrate that Psen1 expression in neural progenitor cells is crucial for cortical development and reveal a novel role for neuroectodermal expression of Psen1 in development of the brain vasculature.


Subject(s)
Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Membrane Proteins/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Cell Movement , Cerebral Cortex/embryology , Cerebral Hemorrhage/genetics , Embryo Loss , Embryo, Mammalian/blood supply , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Intermediate Filament Proteins/genetics , Introns/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nestin , Neurons/pathology , Presenilin-1 , Rats , Stem Cells/pathology , Telencephalon/embryology , Telencephalon/metabolism , Telencephalon/pathology
18.
Exp Neurol ; 196(1): 199-203, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16095592

ABSTRACT

Apical dendritic retraction and axospinous synapse loss in the medial prefrontal cortex (PFC) are structural alterations that result from repeated restraint stress. Such changes in this brain region may be associated with impaired working memory, altered emotionality, and inability to regulate hypothalamic-pituitary adrenal activity, which in turn may underlie stress-related mental illnesses. In the present study, we examined the persistence of these stress-induced dendritic alterations in the medial PFC following the cessation of stress. Animals received either daily restraint stress for a 3-week period and were then allowed to recover for another 3 weeks, restraint stress for 3 or 6 weeks, or no restraint. Following perfusion and fixation, intracellular iontophoretic injections of Lucifer Yellow were performed in layer II/III pyramidal neurons in slices from the medial PFC, and apical and basal dendritic arbors were reconstructed in three dimensions. We observed a significant reduction in apical dendritic length and branch number following 3 or 6 weeks of repeated stress compared to 3-week stress/3-week recovery. These results suggest that stress-induced dendritic plasticity in the medial PFC is reversible and may have implications for the functional recovery of medial PFC function following prolonged psychological stress.


Subject(s)
Dendrites/pathology , Neuronal Plasticity/physiology , Prefrontal Cortex/pathology , Stress, Psychological/physiopathology , Animals , Male , Rats , Rats, Sprague-Dawley , Restraint, Physical/physiology
19.
FASEB J ; 19(6): 659-61, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15650008

ABSTRACT

This study was designed to explore the possibility that caloric restriction (CR) may benefit Alzheimer's disease (AD) by preventing beta-amyloid (Abeta) neuropathology pivotal to the initiation and progression of the disease. We report that a CR dietary regimen prevents Abeta peptides generation and neuritic plaque deposition in the brain of a mouse model of AD neuropathology through mechanisms associated with promotion of anti-amyloidogenic alpha-secretase activity. Study findings support existing epidemiological evidence indicating that caloric intake may influence risk for AD and raises the possibility that CR may be used in preventative measures aimed at delaying the onset of AD amyloid neuropathology.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/pathology , Caloric Restriction , Diet, Carbohydrate-Restricted , Disease Models, Animal , Adipose Tissue , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/analysis , Animals , Aspartic Acid Endopeptidases , Brain Chemistry , Endopeptidases/metabolism , Female , Glucose Tolerance Test , Mice , Organ Size , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
20.
Neurol Res ; 26(5): 573-8, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15265277

ABSTRACT

Age-related and amyloid-induced pathology of the cerebral microvasculature have been implicated as potential contributing factors to the pathogenesis of Alzheimer's disease (AD). The microvasculature plays a crucial role in maintaining brain homeostasis and deterioration of its integrity may have deleterious effects on brain function in AD, possibly leading to neurofibrillary degeneration, plaque formation, and cell loss. Brain vessels possess peculiar anatomical and physiological properties owing to their role in the exchange processes of various substances between blood and brain, which are highly regulated for the maintenance of ionic homeostasis of the neuronal environment. Here we review neuropathological aspects of cortical microvessels in aging and AD in relationship to known cardiovascular risk factors and their possible impact on the cognitive decline seen in late-onset dementia.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Cerebrovascular Circulation , Microcirculation/pathology , Alzheimer Disease/physiopathology , Cerebral Cortex/physiopathology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...