Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
J Alzheimers Dis Rep ; 2(1): 27-39, 2018 Feb 16.
Article in English | MEDLINE | ID: mdl-30480246

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia and there is no successful treatment available. Evidence suggests that fibril formation of the amyloid ß-peptide (Aß) is a major underlying cause of AD, and treatment strategies that reduce the toxic effects of Aß amyloid are sought for. The BRICHOS domain is found in several proteins, including Bri2 (also called integral membrane protein 2B (ITM2B)), mutants of which are associated with amyloid and neurodegeneration, and Bri3 (ITM2C). We have used mouse hippocampal neurons and brain tissues from mice and humans and show Bri3 deposits dispersed on AD plaques. In contrast to what has been shown for Bri2, Bri3 immunoreactivity is decreased in AD brain homogenates compared to controls. Both Bri2 and Bri3 BRICHOS domains interact with Aß40 and Aß42 present in neurons and reduce Aß42 amyloid fibril formation in vitro, but Bri3 BRICHOS is less efficient. These results indicate that Bri2 and Bri3 BRICHOS have different roles in relation to Aß aggregation.

3.
Acta Neuropathol Commun ; 5(1): 43, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28595629

ABSTRACT

Alzheimer disease (AD) is a progressive neurodegenerative disorder and the most common form of dementia. The majority of AD cases are sporadic, while up to 5% are families with an early onset AD (EOAD). Mutations in one of the three genes: amyloid beta precursor protein (APP), presenilin 1 (PSEN1) or presenilin 2 (PSEN2) can be disease causing. However, most EOAD families do not carry mutations in any of these three genes, and candidate genes, such as the sortilin-related receptor 1 (SORL1), have been suggested to be potentially causative. To identify AD causative variants, we performed whole-exome sequencing on five individuals from a family with EOAD and a missense variant, p.Arg1303Cys (c.3907C > T) was identified in SORL1 which segregated with disease and was further characterized with immunohistochemistry on two post mortem autopsy cases from the same family. In a targeted re-sequencing effort on independent index patients from 35 EOAD-families, a second SORL1 variant, c.3050-2A > G, was found which segregated with the disease in 3 affected and was absent in one unaffected family member. The c.3050-2A > G variant is located two nucleotides upstream of exon 22 and was shown to cause exon 22 skipping, resulting in a deletion of amino acids Gly1017- Glu1074 of SORL1. Furthermore, a third SORL1 variant, c.5195G > C, recently identified in a Swedish case control cohort included in the European Early-Onset Dementia (EU EOD) consortium study, was detected in two affected siblings in a third family with familial EOAD. The finding of three SORL1-variants that segregate with disease in three separate families with EOAD supports the involvement of SORL1 in AD pathology. The cause of these rare monogenic forms of EOAD has proven difficult to find and the use of exome and genome sequencing may be a successful route to target them.


Subject(s)
Alzheimer Disease/genetics , Genetic Predisposition to Disease , Genetic Variation , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/genetics , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Brain/pathology , Case-Control Studies , Cohort Studies , Family , Female , Humans , Immunohistochemistry , Male , Middle Aged , Exome Sequencing
4.
J Gerontol A Biol Sci Med Sci ; 72(3): 319-328, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27131040

ABSTRACT

Inflammation plays an important role in Alzheimer's disease (AD) and other neurodegenerative disorders. Although chronic inflammation in later stages of AD is well described, little is known about the inflammatory processes in preclinical or early stages of the disease prior to plaque deposition. In this study, we report that the inflammatory mediator S100A8 is increased with aging in the mouse brain. It is observed as extracellular aggregates, which do not correspond to corpora amylacea. S100A8 aggregation is enhanced in the hippocampi of two different mouse models for amyloid-ß (Aß) overproduction (Tg2576 and TgAPParctic mice). S100A8 aggregates are seen prior the formation of Aß plaques and do not colocalize. In vitro treatment of glial cells from primary cultures with Aß42 resulted in an increased production of S100A8. In parallel, treatment of a neuronal cell line with recombinant S100A8 protein resulted in enhanced Aß42 and decreased Aß40 production. Our results suggest that important inflammatory processes are occurring prior to Aß deposition and the existence of a positive feedback between S100A8 and Aß productions. The possible relevance of aging- or AD-dependent formation of S100A8 aggregates in the hippocampus thus affecting learning and memory processes is discussed.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/physiology , Calgranulin A/physiology , Feedback, Physiological , Hippocampus/pathology , Inflammation/etiology , Protein Aggregation, Pathological , Age Factors , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Animals , Calgranulin A/metabolism , Hippocampus/metabolism , Mice , Mice, Transgenic
5.
Acta Neuropathol Commun ; 4: 22, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26936765

ABSTRACT

INTRODUCTION: The accumulation of insoluble proteins within neurons and glia cells is a pathological hallmark of several neurodegenerative diseases. Abnormal aggregation of the microtubule-associated protein tau characterizes the neuropathology of tauopathies, such as Alzheimer disease (AD), corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP). An impairment of the lysosomal degradation pathway called macroautophagy, hereafter referred to as autophagy, could contribute to the accumulation of aggregated proteins. The role of autophagy in neurodegeneration has been intensively studied in the context of AD but there are few studies in other tauopathies and it is not known if defects in autophagy is a general feature of tauopathies. In the present study, we analysed autophagic and lysosomal markers in human post-mortem brain samples from patients with early-onset familial AD (FAD) with the APP Swedish mutation (APPswe), CBD and PSP and control individuals. RESULTS: FAD, CBD and PSP patients displayed an increase in LC3-positive vesicles in frontal cortex, indicating an accumulation of autophagic vesicles. Moreover, using double-immunohistochemistry and in situ proximity ligation assay, we observed colocalization of hyperphosphorylated tau with the autophagy marker LC3 in FAD, CBD and PSP patients but not in control individuals. Increased levels of the lysosomal marker LAMP1 was detected in FAD and CBD, and in addition Cathepsin D was diffusely spread in the cytoplasm in all tauopathies suggesting an impaired lysosomal integrity. CONCLUSION: Taken together, our results indicate an accumulation of autophagic and lysosomal markers in human brain tissue from patients with primary tauopathies (CBD and PSP) as well as FAD, suggesting a defect of the autophagosome-lysosome pathway that may contribute to the development of tau pathology.


Subject(s)
Alzheimer Disease/pathology , Autophagy/physiology , Brain/pathology , Lysosomes/pathology , Neurodegenerative Diseases/pathology , Supranuclear Palsy, Progressive/pathology , tau Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Brain/metabolism , Female , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Male , Microtubule-Associated Proteins/metabolism , Middle Aged , Nerve Tissue Proteins/metabolism , Postmortem Changes , Sequestosome-1 Protein
6.
Curr Alzheimer Res ; 13(7): 817-30, 2016.
Article in English | MEDLINE | ID: mdl-26825094

ABSTRACT

Several Alzheimer model mice carrying transgenic amyloid precursor protein (APP) with the Swedish mutation have been reported to exhibit spontaneous seizures and/or increased epileptiform EEG activity. The primary cause for the epilepsy phenotype is still under debate. In contrast to mice with APPswe mutation that develop extracellular amyloid plaques, mice with APP Arctic mutation (E693G) have no bias toward ß-secretase cleavage and display intracellular amyloid deposits but not plaques. We conducted a systematic long-term video-EEG recording in three two-week sessions on 21 APParc and 11 wild-type control mice between 3.5 and 8 months of age. Spontaneous seizures were not detected more often in APParc mice than in their wild-type control mice. Long (1 - 5 s) epileptiform discharges were occasionally detected in both APParc and wild-type mice, but short (0.5 - <1 s) epileptiform discharges were more common in APParc mice than in wild-types. However, they were far less frequent than in 6 APPswe/PS1dE9 mice recorded in parallel. In pentylenetetrazole test for seizure susceptibility, APParc mice displayed a shorter latency to sharp-wave discharges than wildtype mice but no increase in seizure duration. These data speak for a relatively mild epilepsy phenotype in APParc mice compared to APPswe mice despite even higher extent of APP overexpression. Thus extracellular amyloid plaques or increased ß-secretase cleavage products appear important for the epilepsy phenotype in APPswe mice.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor/genetics , Brain/physiopathology , Gene Expression Regulation/genetics , Mutation/genetics , Seizures/genetics , Age Factors , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Convulsants/toxicity , Disease Models, Animal , Electroencephalography , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pentylenetetrazole/toxicity , Seizures/etiology , Sleep Stages , Statistics, Nonparametric , Video Recording , Wakefulness
7.
J Neurochem ; 136(3): 497-502, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26500157

ABSTRACT

Accumulation of amyloid ß-peptide (Aß) in the brain is an important event in the pathogenesis of Alzheimer disease. We have used a transgenic mouse model expressing human amyloid precursor protein (APP) with the Arctic mutation to investigate whether Aß deposition is correlated with mitochondrial functions in these animals. We found evidence of mitochondrial dysfunction (i.e., decreased mitochondrial membrane potential, increased production of reactive oxygen species and oxidative DNA damage) at 6 months of age, when the mice showed very mild Aß deposition. More pronounced mitochondrial abnormalities were present in 24-month-old TgAPParc mice with more extensive Aß pathology. This study demonstrates for the first time mitochondrial dysfunction in transgenic mice with a mutation within the Aß peptide (the Arctic APP mutation), and confirms previous studies suggesting that mitochondrial dysfunction and oxidative stress is an early event in the pathogenesis of Alzheimer disease. This study demonstrates mitochondrial dysfunction in transgenic mice with a mutation within the amyloid beta (Aß) peptide (the Arctic amyloid precursor protein (APP) mutation). We found evidence of mitochondrial dysfunction (i.e. decreased mitochondrial membrane potential (MMP), increased production of reactive oxygen species (ROS) and oxidative DNA damage) at 6 months of age, when very mild Aß deposition is present in the mice. Also, the cytochrome c (COX) activity was significantly decreased in mitochondria from transgenic mice at 24 months of age.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Mitochondrial Diseases/genetics , Mutation/genetics , 8-Hydroxy-2'-Deoxyguanosine , Age Factors , Amyloid beta-Peptides/metabolism , Analysis of Variance , Animals , Brain/metabolism , Brain/pathology , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Electron Transport Complex IV/metabolism , Gene Expression Regulation/genetics , Humans , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Reactive Oxygen Species/metabolism
8.
Acta Neuropathol ; 129(4): 541-63, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25676386

ABSTRACT

Alzheimer's disease and other age-related neurodegenerative disorders are associated with deterioration of the noradrenergic locus coeruleus (LC), a probable trigger for mood and memory dysfunction. LC noradrenergic neurons exhibit particularly high levels of somatostatin binding sites. This is noteworthy since cortical and hypothalamic somatostatin content is reduced in neurodegenerative pathologies. Yet a possible role of a somatostatin signal deficit in the maintenance of noradrenergic projections remains unknown. Here, we deployed tissue microarrays, immunohistochemistry, quantitative morphometry and mRNA profiling in a cohort of Alzheimer's and age-matched control brains in combination with genetic models of somatostatin receptor deficiency to establish causality between defunct somatostatin signalling and noradrenergic neurodegeneration. In Alzheimer's disease, we found significantly reduced somatostatin protein expression in the temporal cortex, with aberrant clustering and bulging of tyrosine hydroxylase-immunoreactive afferents. As such, somatostatin receptor 2 (SSTR2) mRNA was highly expressed in the human LC, with its levels significantly decreasing from Braak stages III/IV and onwards, i.e., a process preceding advanced Alzheimer's pathology. The loss of SSTR2 transcripts in the LC neurons appeared selective, since tyrosine hydroxylase, dopamine ß-hydroxylase, galanin or galanin receptor 3 mRNAs remained unchanged. We modeled these pathogenic changes in Sstr2(-/-) mice and, unlike in Sstr1(-/-) or Sstr4(-/-) genotypes, they showed selective, global and progressive degeneration of their central noradrenergic projections. However, neuronal perikarya in the LC were found intact until late adulthood (<8 months) in Sstr2(-/-) mice. In contrast, the noradrenergic neurons in the superior cervical ganglion lacked SSTR2 and, as expected, the sympathetic innervation of the head region did not show any signs of degeneration. Our results indicate that SSTR2-mediated signaling is integral to the maintenance of central noradrenergic projections at the system level, and that early loss of somatostatin receptor 2 function may be associated with the selective vulnerability of the noradrenergic system in Alzheimer's disease.


Subject(s)
Alzheimer Disease/pathology , Locus Coeruleus/metabolism , Norepinephrine/metabolism , Receptors, Somatostatin/metabolism , Age Factors , Aged , Amyloid beta-Peptides/metabolism , Animals , Biogenic Monoamines/metabolism , Carbocyanines/metabolism , Case-Control Studies , Cohort Studies , Female , Gene Expression Regulation/genetics , Humans , Locus Coeruleus/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Neurons/metabolism , Receptors, Somatostatin/genetics , Signal Transduction/physiology , Somatostatin/metabolism , Temporal Lobe/metabolism , Temporal Lobe/pathology , Tyrosine 3-Monooxygenase/metabolism , tau Proteins/metabolism
9.
Acta Neuropathol Commun ; 2: 17, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24517102

ABSTRACT

BACKGROUND: The progressive development of Alzheimer's disease (AD) pathology follows a spatiotemporal pattern in the human brain. In a transgenic (Tg) mouse model of AD expressing amyloid precursor protein (APP) with the arctic (E693G) mutation, pathology spreads along anatomically connected structures. Amyloid-ß (Aß) pathology first appears in the subiculum and is later detected in interconnected brain regions, including the retrosplenial cortex. We investigated whether the spatiotemporal pattern of Aß pathology in the Tg APP arctic mice to interconnected brain structures can be interrupted by destroying neurons using a neurotoxin and thereby disconnecting the neural circuitry. RESULTS: We performed partial unilateral ibotenic acid lesions of the subiculum (first structure affected by Aß pathology) in young Tg APParc mice, prior to the onset of pathology. We assessed Aß/C99 pathology in mice aged up to 6 months after injecting ibotenate into the subiculum. Compared to the brains of intact Tg APP arctic mice, we observed significantly decreased Aß/C99 pathology in the ipsilateral dorsal subiculum, CA1 region of the hippocampus and the retrosplenial cortex; regions connecting to and from the dorsal subiculum. By contrast, Aß/C99 pathology was unchanged in the contralateral hippocampus in the mice with lesions. CONCLUSION: These results, obtained in an animal model of AD, support the notion that Aß/C99 pathology is transmitted between interconnected neurons in AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Functional Laterality , Gene Expression Regulation/physiology , Hippocampus/injuries , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Cell Count , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Gene Expression Regulation/drug effects , Hippocampus/physiology , Humans , Ibotenic Acid/toxicity , Mice , Mice, Transgenic , Mutation
11.
Proc Natl Acad Sci U S A ; 110(19): 7916-21, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23620518

ABSTRACT

It is well-established that subcompartments of endoplasmic reticulum (ER) are in physical contact with the mitochondria. These lipid raft-like regions of ER are referred to as mitochondria-associated ER membranes (MAMs), and they play an important role in, for example, lipid synthesis, calcium homeostasis, and apoptotic signaling. Perturbation of MAM function has previously been suggested in Alzheimer's disease (AD) as shown in fibroblasts from AD patients and a neuroblastoma cell line containing familial presenilin-2 AD mutation. The effect of AD pathogenesis on the ER-mitochondria interplay in the brain has so far remained unknown. Here, we studied ER-mitochondria contacts in human AD brain and related AD mouse and neuronal cell models. We found uniform distribution of MAM in neurons. Phosphofurin acidic cluster sorting protein-2 and σ1 receptor, two MAM-associated proteins, were shown to be essential for neuronal survival, because siRNA knockdown resulted in degeneration. Up-regulated MAM-associated proteins were found in the AD brain and amyloid precursor protein (APP)Swe/Lon mouse model, in which up-regulation was observed before the appearance of plaques. By studying an ER-mitochondria bridging complex, inositol-1,4,5-triphosphate receptor-voltage-dependent anion channel, we revealed that nanomolar concentrations of amyloid ß-peptide increased inositol-1,4,5-triphosphate receptor and voltage-dependent anion channel protein expression and elevated the number of ER-mitochondria contact points and mitochondrial calcium concentrations. Our data suggest an important role of ER-mitochondria contacts and cross-talk in AD pathology.


Subject(s)
Alzheimer Disease/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Receptor Cross-Talk , Amyloid/metabolism , Animals , Brain/metabolism , CHO Cells , Calcium/metabolism , Cell Line, Tumor , Cricetinae , Disease Models, Animal , Gene Knockdown Techniques , Hippocampus/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors , Membrane Microdomains/metabolism , Mice , Mutation , Neurons/metabolism , RNA, Small Interfering/metabolism , Receptors, sigma/metabolism , Subcellular Fractions/metabolism , Vesicular Transport Proteins/metabolism , Sigma-1 Receptor
12.
Neurochem Int ; 60(5): 533-42, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22366649

ABSTRACT

Aggresomes are cytoplasmic inclusions which are localized at the microtubule organizing center (MTOC) as a result of induced proteasome inhibition, stress or over-expression of certain proteins. Aggresomes are linked to the pathogenesis of many neurodegenerative diseases. Here we studied whether amyloid precursor protein (APP), a type-I transmembrane glycoprotein, is localized in aggresomes after exposure to stress condition. Using confocal microscopy we found that APP is located in aggresomes and co-localized with vimentin, γ-tubulin, 20S and ubiquitin at the MTOC in response to proteasome dysfunction. An interaction between vimentin and APP was found after proteasome inhibition suggesting that APP is an additional protein constituent of aggresomes. Suppression of the proteasome system in APP-HEK293 cells overexpressing APP or transfected with APP Swedish mutation caused an accumulation of stable, detergent-insoluble forms of APP containing poly-ubiquitinated proteins. In addition, brain homogenates from transgenic mice expressing human APP with the Arctic mutation demonstrated an interaction between APP and the aggresomal-marker vimentin. These data suggest that malfunctioning of the proteasome system caused by mutation or overexpression of pathological or non-pathological proteins may lead to the accumulation of stable aggresomes, perhaps contributing to the neurodegeneration.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Enzyme Inhibitors/pharmacology , Proteasome Inhibitors , Amyloid beta-Protein Precursor/genetics , Animals , Humans , Mice , Mice, Transgenic , Microscopy, Confocal
13.
Neurobiol Aging ; 33(4): 831.e11-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21880397

ABSTRACT

The Arctic APP mutation (E693G) within the amyloid ß (Aß) domain of amyloid precursor protein (APP) leads to dementia with clinical features similar to Alzheimer's disease (AD), which is believed to be mediated via increased formation of protofibrils. We have generated a transgenic mouse model, TgAPParc, with neuron-specific expression of human amyloid precursor protein with the Arctic mutation (hAPParc), showing mild amyloid pathology with a relatively late onset. Here we performed a detailed analysis of the spatiotemporal progression of neuropathology in homozygous TgAPParc, focusing on intracellular Aß and diffuse Aß aggregates rather than amyloid plaques. We show that the neuropathology in homozygous TgAPParc mice starts with intracellular Aß aggregates, which is followed by diffuse extracellular Aß deposits in subiculum that later expands to brain regions receiving neuronal projections from regions already affected. Together this suggests that the pathology in TgAPParc mice affects interconnected brain regions and may represent a valuable tool to study the spread and progression of neuropathology in Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Hippocampus/metabolism , Mutation/genetics , Peptide Fragments/metabolism , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Female , Hippocampus/pathology , Hippocampus/ultrastructure , Humans , Male , Mice , Mice, Transgenic , Microscopy, Immunoelectron
14.
Neurobiol Aging ; 32(2): 280-92, 2011 Feb.
Article in English | MEDLINE | ID: mdl-19329229

ABSTRACT

The Arctic APP mutation (E693G) leads to dementia with clinical features similar to Alzheimer disease (AD), but little is known about the pathogenic mechanism of this mutation. To address this question, we have generated a transgenic mouse model, TgAPParc, with neuron-specific expression of human APP with the Arctic mutation (hAPParc). Heterozygous mice from two separate founder lines with different levels of expression of hAPParc were analyzed with respect to brain morphology and behavior every 3 months until the age of 18 months. Standard histological stainings and immunohistochemistry using a panel of Aß antibodies showed an age- and dose-dependant progression of amyloid deposition in the brain, starting in the subiculum and spreading to the thalamus. Cognitive behavioral testing revealed deficits in hippocampus-dependent spatial learning and memory in the Barnes maze test. This study demonstrates that the Arctic APP mutation is sufficient to cause amyloid deposition and cognitive dysfunction, and thus the TgAPParc mouse model provides a valuable tool to study the effect of the Arctic mutation in vivo without possible confounding effect of other APP mutations.


Subject(s)
Alanine/genetics , Alzheimer Disease/complications , Alzheimer Disease/pathology , Brain/pathology , Cognition Disorders/etiology , Glycine/genetics , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Cognition Disorders/genetics , Disease Progression , Enzyme-Linked Immunosorbent Assay/methods , Escape Reaction/physiology , Exploratory Behavior/physiology , Humans , Learning Disabilities/etiology , Maze Learning/physiology , Mice , Mice, Transgenic , Mutation/genetics , Peptide Fragments/metabolism , Statistics, Nonparametric
15.
Neurosci Lett ; 476(2): 79-83, 2010 May 31.
Article in English | MEDLINE | ID: mdl-20385205

ABSTRACT

Housing rodents in an enriched environment (EE) induces structural and functional plasticity in the adult brain, including increased dendritic sprouting and number of dendritic spines. However, the molecular mechanisms behind EE-induced brain plasticity remain largely unknown. Circadian rhythm plays an important role in memory processing but the neurobiological mechanisms of how circadian rhythm affects memory and brain plasticity remain controversial. In the current study, we studied the expression of spinophilin, a protein highly enriched in dendritic spines and involved in spine morphology and synaptic plasticity, to examine the effects of EE and circadian rhythm in rats housed in EE for different periods of time. Spinophilin mRNA expression was studied by in situ hybridization and the density of spinophilin immunoreactive puncta was quantified after immunohistochemical staining. Compared to rats living in a deprived environment (DE), we found a transient increase in the density of spinophilin immunoreactive puncta in hippocampus and cortex after 1 week of EE housing and persistent elevations of spinophilin mRNA expression during 1-4 weeks of environmental enrichment. Increased spinophilin expression was found during the light phase of the diurnal cycle, but not the dark phase. Thus, enriched housing altered the diurnal variation in spinophilin mRNA expression, suggesting that circadian modulation is likely to be important for experience dependent plasticity. The current results suggest a possible role for spinophilin in neuronal plasticity induced by environmental enrichment, but further studies are needed to establish a cause-effect relation.


Subject(s)
Cerebral Cortex/metabolism , Dendritic Spines/metabolism , Environment , Hippocampus/metabolism , Microfilament Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , RNA, Messenger/biosynthesis , Animals , Circadian Rhythm , Male , Microfilament Proteins/genetics , Nerve Tissue Proteins/genetics , Rats , Rats, Sprague-Dawley
16.
Brain Res ; 1046(1-2): 137-44, 2005 Jun 07.
Article in English | MEDLINE | ID: mdl-15927552

ABSTRACT

Rodents housed in an enriched environment (EE) show increased neuronal plasticity with enhanced long-term potentiation and memory performance. We report an EE-induced increase in NGFI-A and Krox-20 mRNA expression exclusively during the dark period of the day. In addition, EE-housed rats showed considerable diurnal variation in NGFI-A, Krox-20, and NGFI-B mRNA expression which was absent in single-housed rats. Thus, EE-induced molecular changes are more evident during the dark phase when the rats have higher motor and exploratory activity. This is important to take into account in future studies of molecular mediators of experience-dependent neuronal plasticity.


Subject(s)
Circadian Rhythm/physiology , DNA-Binding Proteins/metabolism , Darkness , Environment , Immediate-Early Proteins/metabolism , Transcription Factors/metabolism , Analysis of Variance , Animals , Brain/metabolism , Circadian Rhythm/genetics , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Early Growth Response Protein 2 , Gene Expression/physiology , Immediate-Early Proteins/genetics , Male , Neuronal Plasticity/physiology , Nuclear Receptor Subfamily 4, Group A, Member 1 , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Transcription Factors/genetics
17.
Neurosci Lett ; 385(2): 173-8, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-15964140

ABSTRACT

Functional recovery after experimental stroke in rats is enhanced by environmental enrichment by stimulating plastic changes in brain regions outside the lesion, but the molecular mechanisms are not known. We investigated the effect of environmental enrichment after focal cerebral ischemia on cognitive recovery and hippocampal gene expression using microarray analysis. Rats placed in enriched environment (EE) for 1 month after middle cerebral artery occlusion (MCAo) showed significantly improved spatial memory in the Morris water maze compared to rats housed alone after MCAo. Microarray analysis suggested several EE-induced differences in neuronal plasticity-related genes, but these changes could not be confirmed by quantitative real-time PCR. This study highlights some of the potential problems associated with gene expression profiling of brain tissues. Further studies at earlier time points and in additional subregions of the brain are of interest in the search for molecular mechanisms behind EE-induced neuronal plasticity after ischemic stroke.


Subject(s)
Brain Ischemia/genetics , Cerebral Infarction/genetics , Environment Design , Hippocampus/metabolism , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Animals , Brain Ischemia/metabolism , Brain Ischemia/therapy , Cerebral Infarction/metabolism , Cerebral Infarction/therapy , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation/genetics , Hippocampus/pathology , Hippocampus/physiopathology , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/therapy , Male , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/therapy , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Recovery of Function/genetics , Time Factors
18.
Eur J Neurosci ; 19(8): 2288-98, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15090055

ABSTRACT

Cognitive impairment is common after ischemic stroke. In rodent stroke models using occlusion of the middle cerebral artery (MCA) this is reflected by impaired spatial memory associated with the size of the ischemic lesion. Housing in an enriched environment enhances brain plasticity and improves recovery of sensorimotor functions after experimental stroke in rats. In this study we report that postischemic housing in an enriched environment also attenuates the long-term spatial memory impairment after MCA occlusion and extinguishes the association between spatial memory and infarct volume. An enriched environment did not significantly alter the expression of selected neuronal plasticity-associated genes 1 month after MCA occlusion, indicating that most of the adaptive changes induced by an enriched environment have already occurred at this time point. We conclude that the attenuated memory impairment induced by environmental enrichment after MCA occlusion provides a useful model for further studies on the neurobiological mechanisms of recovery of cognitive functions after ischemic stroke.


Subject(s)
Brain Ischemia/psychology , Environment , Maze Learning/physiology , Memory Disorders/psychology , Animals , Brain Ischemia/metabolism , Cognition/physiology , Male , Memory Disorders/metabolism , Memory Disorders/prevention & control , Rats , Rats, Sprague-Dawley , Synapsins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...