Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 283
Filter
1.
ACS Nano ; 15(3): 4678-4687, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33666411

ABSTRACT

The progressive accumulation of amyloid-beta (Aß) in specific areas of the brain is a common prelude to late-onset of Alzheimer's disease (AD). Although activation of liver X receptors (LXR) with agonists decreases Aß levels and ameliorates contextual memory deficit, concomitant hypercholesterolemia/hypertriglyceridemia limits their clinical application. DMHCA (N,N-dimethyl-3ß-hydroxycholenamide) is an LXR partial agonist that, despite inducing the expression of apolipoprotein E (main responsible of Aß drainage from the brain) without increasing cholesterol/triglyceride levels, shows nil activity in vivo because of a low solubility and inability to cross the blood brain barrier. Herein, we describe a polymer therapeutic for the delivery of DMHCA. The covalent incorporation of DMHCA into a PEG-dendritic scaffold via carboxylate esters produces an amphiphilic copolymer that efficiently self-assembles into nanometric micelles that exert a biological effect in primary cultures of the central nervous system (CNS) and experimental animals using the intranasal route. After CNS biodistribution and effective doses of DMHCA micelles were determined in nontransgenic mice, a transgenic AD-like mouse model of cerebral amyloidosis was treated with the micelles for 21 days. The benefits of the treatment included prevention of memory deterioration and a significant reduction of hippocampal Aß oligomers without affecting plasma lipid levels. These results represent a proof of principle for further clinical developments of DMHCA delivery systems.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Liver X Receptors , Mice , Mice, Transgenic , Polymers , Tissue Distribution
2.
J Prev Alzheimers Dis ; 6(1): 2-15, 2019.
Article in English | MEDLINE | ID: mdl-30569080

ABSTRACT

Scientific evidence collected over the past 4 decades suggests that a loss of cholinergic innervation in the cerebral cortex of patients with Alzheimer's disease is an early pathogenic event correlated with cognitive impairment. This evidence led to the formulation of the "Cholinergic Hypothesis of AD" and the development of cholinesterase inhibitor therapies. Although approved only as symptomatic therapies, recent studies suggest that long-term use of these drugs may also have disease-modifying benefits. A Cholinergic System Workgroup reassessed the role of the cholinergic system on AD pathogenesis in light of recent data, including neuroimaging data charting the progression of neurodegeneration in the cholinergic system and suggesting that cholinergic therapy may slow brain atrophy. Other pathways that contribute to cholinergic synaptic loss and their effect on cognitive impairment in AD were also reviewed. These studies indicate that the cholinergic system as one of several interacting systems failures that contribute to AD pathogenesis.


Subject(s)
Alzheimer Disease , Cholinergic Agents/therapeutic use , Cholinergic Neurons/pathology , Cholinergic Neurons/physiology , Translational Research, Biomedical , Aging/physiology , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Brain/pathology , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Dementia/pathology , Dementia/physiopathology , Humans
3.
Transl Psychiatry ; 7(8): e1190, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28763060

ABSTRACT

Lithium is first-line therapy for bipolar affective disorder and has recently been shown to have protective effects in populations at risk for Alzheimer's disease (AD). However, the mechanism underlying this protection is poorly understood and consequently limits its possible therapeutic application in AD. Moreover, conventional lithium formulations have a narrow therapeutic window and are associated with a severe side effect profile. Here we evaluated a novel microdose formulation of lithium, coded NP03, in a well-characterized rat model of progressive AD-like amyloid pathology. This formulation allows microdose lithium delivery to the brain in the absence of negative side effects. We found that NP03 rescued key initiating components of AD pathology, including inactivating GSK-3ß, reducing BACE1 expression and activity, and reducing amyloid levels. Notably, NP03 rescued memory loss, impaired CRTC1 promoter binding of synaptic plasticity genes and hippocampal neurogenesis. These results raise the possibility that NP03 be of therapeutic value in the early or preclinical stages of AD.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Hippocampus/drug effects , Lithium/administration & dosage , Memory Disorders/drug therapy , Memory/drug effects , Neuroprotective Agents/administration & dosage , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Female , Hippocampus/metabolism , Hippocampus/pathology , Lithium/therapeutic use , Male , Memory Disorders/metabolism , Memory Disorders/pathology , Neurogenesis/drug effects , Neuroprotective Agents/therapeutic use , Rats , Rats, Transgenic
4.
Neurodegener Dis ; 10(1-4): 104-7, 2012.
Article in English | MEDLINE | ID: mdl-22261363

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) neuropathology likely begins decades before clinical symptoms are manifested. Investigations on the early stages of the amyloid pathology are crucial for the discovery of diagnostic biomarkers or new therapeutic targets. Our transgenic (tg) animal models are most suitable to study early AD pathological events, as the pathology evolves in a well-staged manner, starting with intracellular Aß accumulation and ending with plaque deposition. OBJECTIVE: To determine the occurrence of key inflammatory markers and to look for signs of nerve growth factor (NGF) dysmetabolism at preplaque and postplaque stages in tg models of AD-like amyloid pathology and in human AD brains. METHODS: We used our own tg lines (mice and rat), high-quality human brain material and applied neurochemical and immunohistochemical experimental approaches. RESULTS: In both tg models, we observed an intracellular accumulation of oligomeric Aß in cortical and hippocampal pyramidal neurons. This coincided with an upregulation of key inflammatory markers (iNOS, MHCII, COX-2) and a recruitment of microglia towards Aß-burdened neurons. Using human AD brains, we showed alterations in the NGF metabolic pathway, which were mirrored in our tg rat model at early and late stages of amyloid plaque generation. CONCLUSION: A proinflammatory process and, consequently, the deregulation of the NGF metabolic pathway could be amongst the earliest pathological events in the progression of AD.


Subject(s)
Alzheimer Disease/pathology , Central Nervous System/metabolism , Inflammation/chemically induced , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Genetically Modified , Cyclooxygenase 2/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Mutation/genetics , Nerve Growth Factor/metabolism , Nitric Oxide Synthase Type II/metabolism , Protein Precursors/metabolism , Rats
5.
Curr Alzheimer Res ; 8(2): 164-74, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21345170

ABSTRACT

The occurrence of a plaque-dependent inflammation in Alzheimer's disease has been extensively documented in both human specimens and transgenic models of the disease. Since insoluble plaques are present in AD patients from early preclinical stages of the pathology, the point at which neuroinflammation first occurs in the progression of the AD pathology is still unknown. In this review we discuss the clinical and experimental evidence for the occurrence of inflammation in preclinical, asymptomatic phases of the progression of the AD pathology. In particular, we discuss the evidence from different transgenic models suggesting that a pro-inflammatory process might even be initiated prior to plaque deposition. The factors responsible for the early, pre-plaque inflammation are reviewed, with particular emphasis on the role of soluble Aß oligomers. Furthermore, we analyze the consequences of the microglial activation and the deregulation of NGF metabolism, in the context of the earliest amyloid pathology. Finally, we propose MMP-9 as a promising biomarker for signalling early stages of an ongoing CNS inflammation.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Cognition Disorders/immunology , Inflammation/complications , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Cognition Disorders/metabolism , Cognition Disorders/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Microglia/metabolism , Microglia/pathology , Nerve Growth Factor/metabolism
6.
Curr Alzheimer Res ; 8(1): 4-23, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21143159

ABSTRACT

At diagnosis, Alzheimer's disease (AD) brains are extensively burdened with plaques and tangles and display a degree of synaptic failure most likely beyond therapeutic treatment. It is therefore crucial to identify early pathological events in the progression of the disease. While it is not currently feasible to identify and study early, pre-clinical stages of AD, transgenic (Tg) models offer a valuable tool in this regard. Here we investigated cognitive, structural and biochemical CNS alterations occurring in our newly developed McGill-Thyl-APP Tg mice (over-expressing the human amyloid precursor protein with the Swedish and Indiana mutations) prior to extracellular plaque deposition. Pre-plaque, 3-month old Tg mice already displayed cognitive deficits concomitant with reorganization of cortical cholinergic pre-synaptic terminals. Conformational specific antibodies revealed the early appearance of intracellular amyloid ß (Aß)-oligomers and fibrillar oligomers in pyramidal neurons of cerebral cortex and hippocampus. At the same age, the cortical levels of insulin degrading enzyme -a well established Aß-peptidase, were found to be significantly down-regulated. Our results suggest that, in the McGill-Thy1-APP Tg model, functional, structural and biochemical alterations are already present in the CNS at early, pre-plaque stages of the pathology. Accumulation of intraneuronal neurotoxic Aß-oligomers (possibly caused by a failure in the clearance machinery) is likely to be the culprit of such early, pre-plaque pathology. Similar neuronal alterations might occur prior to clinical diagnosis in AD, during a yet undefined 'latent' stage. A better understanding of such pre-clinical AD might yield novel therapeutic targets and or diagnostic tools.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Disease Models, Animal , Age Factors , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Cerebellum/metabolism , Cerebral Cortex/metabolism , Cognition Disorders/etiology , Drug Evaluation, Preclinical , Gene Expression Regulation/genetics , Hippocampus/metabolism , Humans , Maze Learning/physiology , Mice , Mice, Transgenic , Mutation/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Peptide Fragments/metabolism , Phosphopyruvate Hydratase/metabolism , Recognition, Psychology/physiology , Vesicular Acetylcholine Transport Proteins/metabolism
7.
Neurodegener Dis ; 7(1-3): 96-8, 2010.
Article in English | MEDLINE | ID: mdl-20173335

ABSTRACT

BACKGROUND: Intracellular accumulation of beta-amyloid (Abeta) is one of the early features in the neuropathology of Alzheimer's disease (AD) and Down's syndrome. This can be reproduced in cell and transgenic animal models of the AD-like amyloid pathology. In a transgenic rat model, our lab has previously shown that the intracellular accumulation of Abeta is sufficient to provoke cognitive impairments and biochemical alterations in the cerebral cortex and hippocampus in the absence of amyloid plaques. OBJECTIVE: To investigate an early, pre-plaque inflammatory process in AD-like transgenic models and establish whether the neurotoxic effects of Abeta oligomers and proinflammatory responses can be arrested with minocycline. METHODS: For these studies, we used naïve mice and transgenic animal models of the AD-like amyloid pathology and applied neurochemical, immunohistochemical and behavioral experimental approaches. RESULTS: In the early stages of the AD-like amyloid pathology, intracellular Abeta oligomers accumulate within neurons of the cerebral cortex and hippocampus. Coincidental with this, behavioral impairments occur prior to the appearance of amyloid plaques, together with an upregulation of MHC-II, i-NOS and COX-2, well-known proinflammatory markers. Treatment with minocycline corrected behavioral impairments, lowered inflammatory markers and levels of Abeta trimers. CONCLUSION: A pharmacological approach targeting the early neuroinflammatory effects of Abeta might be a promising strategy to prevent or delay the onset of AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Inflammation/etiology , Alzheimer Disease/complications , Animals , Animals, Genetically Modified , Cognition Disorders/etiology , Disease Models, Animal , Inflammation/pathology , Mice , Rats
8.
Neuroscience ; 159(2): 896-907, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19105974

ABSTRACT

Age-related cognitive impairments are associated with structural and functional changes in the cerebral cortex. We have previously demonstrated in the rat that excitatory and inhibitory pre- and postsynaptic changes occur with respect to age and cognitive status; however, in aged cognitively impaired animals, we have shown a significant imbalance in postsynaptic markers of excitatory versus inhibitory synapses, using markers of excitatory versus inhibitory neurotransmitter-related scaffolding proteins [postsynaptic density-95 (PSD95)/synapse associated protein-90 (SAP90) and gephyrin, respectively]. The present study focuses on whether the expression of various excitatory and inhibitory postsynaptic proteins is affected by ageing and cognitive status. Thus, aged animals were segregated into aged cognitively impaired (AI) and aged cognitively unimpaired (AU) groups using the Morris water maze. We applied Western immunoblotting to reveal the expression patterns of a number of relevant excitatory and inhibitory receptors in the prefrontal and parietal cortices of young (Y), AU and AI animals, and performed semi-quantitative analyses to statistically tabulate changes among the three animal groups. A significant increase in the inhibitory postsynaptic scaffold protein, gephyrin, was observed in the parietal cortex of AI animals. Similarly, an increase in GABA(A) receptor subunit alpha1 was observed in the parietal cortex of AI animals. An increase in the excitatory N-methyl-d-aspartate receptor subunit N-methyl-d-aspartate receptor 1 expression was observed in the parietal cortex of AI animals, whereas a significant decrease in AMPA receptor subunit glutamate receptor 2 expression was found in the prefrontal cortex of AI animals. Finally, the excitatory, postsynaptic neuronal cell-adhesion receptor, neuroligin-1, was found to be significantly increased in both the prefrontal and parietal cortical areas of AI animals.


Subject(s)
Aging/physiology , Cerebral Cortex/metabolism , Cognition Disorders/metabolism , Gene Expression Regulation/physiology , Synapses/metabolism , Age Factors , Analysis of Variance , Animals , Behavior, Animal/physiology , Carrier Proteins/metabolism , Cell Adhesion Molecules, Neuronal , Cerebral Cortex/cytology , Male , Maze Learning/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Rats , Rats, Inbred F344 , Reaction Time/physiology , Receptors, AMPA/metabolism , Receptors, GABA-A/metabolism
9.
Curr Alzheimer Res ; 4(4): 351-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17908036

ABSTRACT

Forebrain cholinergic neurons are highly dependent on nerve growth factor (NGF) for phenotype maintenance. We have established that in addition to "target-derived" NGF neurotrophic stimulation, cholinergic neurons also respond dose-dependently, to intra-parenchymal NGF administration in the somato-dendritic region of the nucleus Basalis, thus illustrating the potential of alternative reparative therapies which would by-pass the undesirable effects of diffuse neurotrophin application. Moreover, our lab has also observed that the steady-state number of cortical cholinergic synapses is dependent on continuous NGF supply, as anti-NGF monoclonal antibodies and TrkA receptor antagonists deplete pre-existing cholinergic bouton numbers. Furthermore, the application of either NGF or TrkA NGF-mimetic agonists successfully rescues the age-dependent loss of cortical cholinergic boutons in aged-impaired rats. The vulnerability of the cortical cholinergic system has also been demonstrated in transgenic animal models of the Alzheimer's disease (AD) amyloid pathology. It is of interest to note however, that an up-regulation of cholinergic presynaptic boutons has been observed in certain transgenic mouse models prior to plaque formation. This observation is similar to the visibly increased immunoreactivity of cortical and hippocampal choline acetyltransferase (ChAT) fibers in patients with Mild Cognitive Impairment (MCI). A series of ex-vivo experiments conducted by our group have demonstrated that contrary to popular belief, proNGF, as opposed to mature NGF, is released from the cerebral cortex in an activity-dependent manner. In addition, proNGF appears to be released with a series of pro-enzymes and enzymes, which are involved in its subsequent maturation to NGF and degradation in the extracellular space. Given that proNGF is known to be upregulated in AD patients a dysregulation in the maturation or degradation of mature NGF might explain the preferential vulnerability of the cholinergic system in the AD pathology.


Subject(s)
Acetylcholine/metabolism , Aging , Alzheimer Disease/complications , Cognition Disorders/metabolism , Nerve Growth Factor/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cognition Disorders/etiology , Cognition Disorders/pathology , Humans
10.
J Neurochem ; 103(1): 216-28, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17587310

ABSTRACT

The beta-amyloid peptide (Abeta) is considered responsible for the pathogenesis of Alzheimer's disease. Despite the magnitude of reports describing a neurotoxic role of extracellular Abeta, the role for intracellular Abeta (iAbeta) has not been elucidated. We previously demonstrated that in rat pheochromocytoma cells expression of moderate levels of Abeta results in the up-regulation of phospho-extracellular signal-regulated kinases (ERK1)/2 along with an elevation of cyclic AMP-response element (CRE)-regulated gene expression; however, the effect of high intracellular levels of Abeta were not examined. Towards this goal we generated constructs that endogenously produce different expression levels of iAbeta in a human cell line. We show a bimodal response to Abeta in a neural human cell line. A moderate increase of endogenous Abeta up-regulates certain cyclic AMP-response element-binding protein (CREB) responsive genes such as presenilin 1, presenilin 2, brain-derived neurotrophic factor, and mRNA and protein levels by CREB activation and Synapsin 1 nuclear translocation. On the other hand, high-loads of iAbeta resulted in sustained hyper-phosphorylation of CREB that did not translocate to the nucleus and did not stimulate activation of CRE-regulated gene expression. Our study suggests that variations in levels of iAbeta could influence signaling mechanisms that lead to phosphorylation of CREB, its nuclear translocation and CRE-regulated genes involved in production of Abeta and synaptic plasticity in opposite directions.


Subject(s)
Amyloid beta-Peptides/metabolism , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Intracellular Fluid/metabolism , Active Transport, Cell Nucleus , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Gene Expression Regulation , Gene Transfer Techniques , Humans , Mutation , PC12 Cells , Phosphorylation , Rats
11.
Neuroscience ; 135(4): 1193-202, 2005.
Article in English | MEDLINE | ID: mdl-16181736

ABSTRACT

Extracellular-regulated kinases play a fundamental role in several neuroplasticity processes. In order to test whether endogenous beta-amyloid peptides play a role in the activation of extracellular-regulated kinase, we investigated the Rap1-extracellular-regulated kinase pathway in PC12 cells expressing human beta-amyloid precursor protein containing familial Alzheimer's disease mutations. In PC12 cells transfected with mutant human beta-amyloid precursor proteins that lead to higher levels of endogenous beta-amyloid, we observed an up-regulation of phospho-extracellular-regulated kinase and higher levels of activity-induced cAMP response element-directed gene expression. These results suggest that moderate levels of endogenous beta-amyloid peptides stimulate cAMP response element-directed gene expression. This stimulation was via a Rap1/MEK/extracellular-regulated kinase signaling pathway, as it was blocked by inhibition of Rap1 and MEK activities, and it requires beta-amyloid precursor protein cleavage at the gamma-site as it was abolished by a gamma-secretase inhibitor. Interestingly, in agreement with the previous observations, micromolar levels of extracellular fibrillar beta-amyloid blocked the cAMP response element-regulated gene expression stimulated by potassium and forskolin. This indicates that beta-amyloid can provoke different responses on cAMP response element-directed gene expression, such that low beta-amyloid levels may play a physiological role favoring synaptic plasticity under normal conditions while it would inhibit this mechanism under pathological conditions.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression , Models, Biological , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases , Blotting, Western , Cyclic AMP Response Element-Binding Protein/drug effects , Endopeptidases/drug effects , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Kinase Kinases/metabolism , Mutation , PC12 Cells , Phosphorylation , Rats , Transfection , rap1 GTP-Binding Proteins/metabolism
12.
Neuroscience ; 129(3): 583-92, 2004.
Article in English | MEDLINE | ID: mdl-15541880

ABSTRACT

The pathological significance of intracellular Abeta accumulation in vivo is not yet fully understood. To address this, we have studied transgenic rats expressing Alzheimer's-related transgenes that accumulate Abeta intraneuronally in the cerebral and hippocampal cortices but do not develop extracellular amyloid plaques. In these rats, the presence of intraneuronal Abeta is sufficient to provoke up-regulation of the phosphorylated form of extracellular-regulated kinase (ERK) 2 and its enzymatic activity in the hippocampus while no changes were observed in the activity or phosphorylation status of other putative tau kinases such as p38, glycogen synthase kinase 3, and cycline-dependent kinase 5. The increase in active phospho-ERK2 was accompanied by increased levels of tau phosphorylation at S396 and S404 ERK2 sites and a decrease in the phosphorylation of the CREB kinase p90RSK. In a water maze paradigm, male transgenic rats displayed a mild spatial learning deficit relative to control littermates. Our results suggest that in the absence of plaques, intraneuronal accumulation of Abeta peptide correlates with the initial steps in the tau-phosphorylation cascade, alterations in ERK2 signaling and impairment of higher CNS functions in male rats.


Subject(s)
Amyloid beta-Peptides/metabolism , Memory Disorders/physiopathology , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , tau Proteins/metabolism , Amyloid beta-Peptides/genetics , Analysis of Variance , Animals , Animals, Genetically Modified , Animals, Newborn , Behavior, Animal , Blotting, Western/methods , Brain/cytology , Humans , Immunohistochemistry/methods , Maze Learning/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Memory Disorders/genetics , Phosphorylation , Presenilin-1 , Rats , Rats, Wistar , Reaction Time/physiology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/genetics
13.
J Alzheimers Dis ; 6(4): 421-31; discussion 443-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15345813

ABSTRACT

Alzheimer's disease (AD) studies typically focus on the extracellular impact of the amyloid-beta (Abeta) protein, however recent findings also implicate intracellular Abeta (iAbeta) accumulation in the disease's molecular neuropathology. In a double mutant transgenic rat model (AbetaPP and PS1 mutations, UKUR25), stably expressing intracellular human Abeta fragments in an environment devoid of both amyloid plaques and neurofibrillary tangles, we investigated the impact of iAbeta burden on both the incidence and relative cross sectional areas of the Golgi apparatus, lysosomes and lipofuscin bodies. Pyramidal cells within the hippocampus and neocortex of both transgenic and non-transgenic age matched controls were compared. This comparison revealed a significant increase in both the proportional area occupied by Golgi apparatus elements as well as in the mean individual cross sectional area of Golgi compartments in the hippocampus of transgenic rats as compared to controls. Elevated lysosome and lipofuscin elements in the hippocampi of transgenic rats were observed, as was an increase in the mean individual, cross sectional area of lipofuscin bodies in the cortex of transgenic rats as compared to controls. These findings support the hypothesis that intracellular Abeta accumulation not only has an impact on subcellular compartments but also potentially contributes to the neuronal cell pathology observed in AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Intracellular Space/metabolism , Neocortex/metabolism , Neocortex/pathology , Neurons/metabolism , Neurons/pathology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/immunology , Animals , Animals, Genetically Modified , Antibodies, Monoclonal/immunology , Golgi Apparatus/metabolism , Lipofuscin/metabolism , Lysosomes/metabolism , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Rats
14.
Neurosci Lett ; 353(2): 143-7, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14664921

ABSTRACT

While the cholinergic depletion in Alzheimer's disease (AD) has been known for some time, a definitive involvement of other neurotransmitter systems has been somewhat more elusive. Our study demonstrates a clear involvement of both glutamatergic and, to a lesser extent, GABAergic neurons in an early onset transgenic mouse model of AD-like amyloid pathology. Immunohistochemical staining and subsequent quantification has revealed a statistically significant increased density of glutamatergic and GABAergic presynaptic boutons in both the plaque free and plaque adjacent cortical neuropile areas of transgenic mice as compared to non-transgenic controls. Furthermore, amyloid plaque size was shown to have a statistically significant effect on the relative area occupied by dystrophic glutamatergic neurites in the peri-plaque neuropile. These findings support our hypothesis that the amyloid pathology progresses in a time and neurotransmitter specific manner, first in the cholinergic system which appears to be most vulnerable, followed by the glutamatergic presynaptic boutons and finally the somewhat more resilient GABAergic terminals.


Subject(s)
Glutamic Acid/metabolism , Neurites/pathology , Plaque, Amyloid/pathology , Presynaptic Terminals/pathology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Disease Models, Animal , Image Processing, Computer-Assisted , Immunohistochemistry , Mice , Mice, Transgenic , Neuropil/metabolism , Neuropil/pathology , Presynaptic Terminals/metabolism , gamma-Aminobutyric Acid/metabolism
15.
Neuroscience ; 121(2): 421-32, 2003.
Article in English | MEDLINE | ID: mdl-14522000

ABSTRACT

A previous study in our laboratory, involving early stage, amyloid pathology in 8-month-old transgenic mice, demonstrated a selective loss of cholinergic terminals in the cerebral and hippocampal cortices of doubly transgenic (APP(K670N,M671L)+PSl(M146L)) mice, an up-regulation in the single mutant APP(K670N,M671L) mice and no detectable change in the PSl(M146L) transgenics [J Neurosci 19 (1999) 2706]. The present study investigates the impact of amyloid plaques on synaptophysin and vesicular acetylcholine transporter (VAChT) immunoreactive bouton numbers in the frontal cortex of the three transgenic mouse models previously described. When compared as a whole, the frontal cortices of transgenic and control mice show no observable differences in the densities of synaptophysin-immunoreactive boutons. An individual comparison of layer V of the frontal cortex, however, shows a significant increase in density in transgenic models. Analysis of the cholinergic system alone shows significant alterations in the VAChT-immunoreactive bouton densities as evidenced by an increased density in the single (APP(K670N,M671L)) transgenics and a decreased density in the doubly transgenics (APP(K670N,M671L)+PSl(M146L)). In investigating the impact of plaque proximity on bouton density at early stages of the amyloid pathology in our doubly (APP(K670N,M671L)+PSl(M146L)) transgenic mouse line, we observed that plaque proximity reduced cholinergic pre-synaptic bouton density by 40%, and yet increased synaptophysin-immunoreactive pre-synaptic bouton density by 9.5%. Distance from plaques (up to 60 microm) seemed to have no effect on bouton density; however a significant inverse relationship was visible between plaque size and cholinergic pre-synaptic bouton density. Finally, the number of cholinergic dystrophic neurites surrounding the truly amyloid, Thioflavin-S(+) plaque core, was disproportionately large with respect to the incidence of cholinergic boutons within the total pre-synaptic bouton population. Confocal and electron microscopic observations confirmed the preferential infiltration of dystrophic cholinergic boutons into fibrillar amyloid aggregates. We therefore hypothesize that extracellular Abeta aggregation preferentially affects cholinergic terminations prior to progression onto other neurotransmitter systems. This is supported by the observable presence of non-cholinergic sprouting, which may be representative of impending neuritic degeneration.


Subject(s)
Acetylcholine/metabolism , Alzheimer Disease/metabolism , Frontal Lobe/metabolism , Membrane Transport Proteins , Neuropeptides , Plaque, Amyloid/metabolism , Presynaptic Terminals/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Cell Count , Cerebral Cortex/metabolism , Disease Models, Animal , Hippocampus/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , Neuropil/metabolism , Presenilin-1 , Presynaptic Terminals/ultrastructure , Synaptophysin/metabolism , Vesicular Biogenic Amine Transport Proteins
16.
Neuroscience ; 108(1): 157-66, 2001.
Article in English | MEDLINE | ID: mdl-11738139

ABSTRACT

Research using animal models of neuropathic pain has revealed sympathetic sprouting onto dorsal root ganglion cells. More recently, sensory fibre sprouting onto dorsal root ganglion cells has also been observed. Previous work in our laboratory demonstrated persistent sympathetic fibre sprouting in the skin of the rat lower lip following sensory denervation of this region. Therefore, we applied immunocytochemistry to determine the effects of sympathectomies on the terminal fields of sensory fibres. The superior cervical ganglia were removed bilaterally and the effects on the innervation of the skin of the rat lower lip were observed 1, 2, 3, 4, 6 and 8 weeks post-surgery. Substance P and dopamine-beta-hydroxylase immunoreactivities were used to identify a subset of sensory and sympathetic fibres, respectively. We also assessed neurokinin-1 receptor immunoreactivity. Quantitative data was obtained with the aid of an image analysis system. In controls, the epidermis and upper dermis were innervated by substance P-immunoreactive fibres only and upper dermal blood vessels possessed the highest density of neurokinin-1 receptor immunoreactivity. Blood vessels in the lower dermis were innervated by both substance P- and dopamine-beta-hydroxylase-immunoreactive fibres. Following sympathectomies, substance P-immunoreactive fibres in the epidermis and upper dermis were more intensely labelled only 1 and 2 weeks post-surgery when compared to sham controls. The length of substance P-immunoreactive fibres in this region was also increased only on the second week. Neurokinin-1 receptor immunoreactivity in the upper dermis was slightly decreased 1 and 2 weeks post-surgery. In the lower dermis, substance P-immunoreactive fibres associated with blood vessels were more intensely labelled only 1 and 2 weeks post-surgery, and at all post-surgical time points studied, blood vessels in this region were devoid of dopamine-beta-hydroxylase-immunoreactive fibres. The length of substance P-immunoreactive fibres was increased from the first to the third week post-surgery in the lower dermis. These results indicate that sympathectomies lead to transient changes in substance P-immunoreactive fibre innervation and neurokinin-1 receptor expression in rat lower lip skin. The effects are most prominent in the lower dermis probably due to a greater local concentration of nerve growth factor in this region. The plasticity of the interactions between sensory and sympathetic fibres may prove important in the regulation of skin microcirculation and in the generation of painful sensations under normal conditions or following peripheral nerve injuries.


Subject(s)
Nerve Fibers/physiology , Neuronal Plasticity/physiology , Neurons, Afferent/physiology , Skin/innervation , Substance P/metabolism , Sympathectomy , Animals , Dermis/innervation , Dopamine beta-Hydroxylase/metabolism , Epidermis/innervation , Immunohistochemistry , Male , Rats , Rats, Wistar , Receptors, Neurokinin-1/metabolism , Time Factors
17.
Neuroscience ; 105(2): 277-85, 2001.
Article in English | MEDLINE | ID: mdl-11672595

ABSTRACT

This study addresses the issue of whether cholinergic varicosities in the cerebral cortex establish 'classical synapses' or whether they communicate with their targets non-synaptically by 'volume transmission'. Most recent studies in the neocortex have suggested that acetylcholine acts non-synaptically, however in the present study we provide ultrastructural evidence that suggests synaptic mechanisms prevail. This conclusion is based upon our ultrastructural observations that cholinergic boutons--as revealed by immunoreactivity for the specific cholinergic market, vesicular acetylcholine transporter--establish a high percentage of classical synapses in layer V of the rat parietal cortex. Furthermore, the combination of this approach with the intracellular labeling of large pyramidal neurons on slice preparations revealed significant incidences of cholinergic contacts abutting preferentially on dendritic shafts. Finally, we have gathered information suggesting that cholinergic boutons undergo atrophy with aging which could be related to the well-known cholinergic and cognitive decline. These results illustrate that the cholinergic terminations in the neocortex establish proper synaptic connections and that they experience important age-dependent atrophy.


Subject(s)
Acetylcholine/metabolism , Aging/pathology , Cerebral Cortex/ultrastructure , Cholinergic Fibers/ultrastructure , Dendrites/ultrastructure , Membrane Transport Proteins , Presynaptic Terminals/ultrastructure , Pyramidal Cells/ultrastructure , Vesicular Transport Proteins , Aging/metabolism , Animals , Atrophy/metabolism , Atrophy/pathology , Carrier Proteins/metabolism , Cerebral Cortex/metabolism , Cholinergic Fibers/metabolism , Dendrites/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Microscopy, Electron , Patch-Clamp Techniques , Presynaptic Terminals/metabolism , Pyramidal Cells/metabolism , Rats , Rats, Inbred F344 , Vesicular Acetylcholine Transport Proteins
18.
J Comp Neurol ; 432(4): 466-80, 2001 Apr 16.
Article in English | MEDLINE | ID: mdl-11268009

ABSTRACT

Cutaneous antidromic vasodilatation and plasma extravasation, two phenomena that occur in neurogenic inflammation, are partially blocked by substance P (SP) receptor antagonists and are known to be mediated in part by mast cell-released substances, such as histamine, serotonin, and nitric oxide. In an attempt to provide a morphological substrate for the above phenomena, we applied light and electron microscopic immunocytochemistry to investigate the pattern of SP innervation of blood vessels and its relationship to mast cells in the skin of the rat lower lip. Furthermore, we examined the distribution of SP (neurokinin-1) receptors and their relationship to SP-immunoreactive (IR) fibers. Our results confirmed that SP-IR fibers are found in cutaneous nerves and that terminal branches are observed around blood vessels and penetrating the epidermis. SP-IR fibers also innervated hair follicles and sebaceous glands. At the ultrastructural level, SP-IR varicosities were observed adjacent to arterioles, capillaries, venules, and mast cells. The varicosities possessed both dense core vesicles and agranular synaptic vesicles. We quantified the distance between SP-IR varicosities and blood vessel endothelial cells. SP-IR terminals were located within 0.23-5.99 microm from the endothelial cell layer in 82.7% of arterioles, in 90.2% of capillaries, and in 86.9% of venules. Although there was a trend for SP-IR fibers to be located closer to the endothelium of venules, this difference was not significant. Neurokinin-1 receptor (NK-1r) immunoreactivity was most abundant in the upper dermis and was associated with the wall of blood vessels. NK-1r were located in equal amounts on the walls of arterioles, capillaries, and venules that were innervated by SP-IR fibers. The present results favor the concept of a participation of SP in cutaneous neurogenic vasodilatation and plasma extravasation both by an action on blood vessels after binding to the NK-1r and by causing the release of substances from mast cells after diffusion through the connective tissue.


Subject(s)
Lip/metabolism , Nerve Fibers/metabolism , Receptors, Neurokinin-1/metabolism , Skin/metabolism , Substance P/metabolism , Animals , Blood Vessels/cytology , Blood Vessels/innervation , Blood Vessels/metabolism , Epidermal Cells , Epidermis/innervation , Epidermis/metabolism , Hair Follicle/cytology , Hair Follicle/innervation , Hair Follicle/metabolism , Lip/blood supply , Lip/innervation , Male , Mast Cells/cytology , Mast Cells/metabolism , Microscopy, Electron , Nerve Fibers/ultrastructure , Rats , Rats, Wistar , Sebaceous Glands/cytology , Sebaceous Glands/innervation , Sebaceous Glands/metabolism , Skin/cytology , Skin/innervation
19.
J Neurosci ; 20(22): 8596-606, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11069968

ABSTRACT

Reduction in both presynaptic and postsynaptic structures in the aging neocortex may significantly affect functional synaptic properties in this area. To directly address this issue, we combined whole-cell patch-clamp recording of spontaneously occurring postsynaptic currents (PSCs) with morphological analysis of layer V pyramidal neurons in the parietal cortex of young adult (1- to 2-month-old) and aged (28- to 37-month-old) BN x F344 F(1) hybrid rats. Analysis of spontaneous PSCs was used to contrast functional properties of basal synaptic input with structural alterations in the dendritic tree of pyramidal neurons and density of terminals in contact with these cells. We observed significant changes in a number of morphological parameters of pyramidal neurons in aged rats. These include smaller cell body size and fewer basal dendritic branches (but not of oblique dendrites and dendritic tufts) and spines. Ultrastructural analysis also revealed a lower density of presynaptic terminals per unit length of postsynaptic membrane of labeled pyramidal neurons in the aged brain. This reduction in both presynaptic and postsynaptic elements was paralleled by a significant decrease in frequency of tetrodotoxin-insensitive miniature (action potential-independent) PSCs (mPSCs). The frequency of excitatory and inhibitory mPSCs was reduced to the same extent. In contrast, no significant change was observed in the frequency of spontaneous PSCs recorded in absence of tetrodotoxin (sPSCs), indicating an increase in action potential-dependent (frequency(sPSCs) - frequency(mPSCs)) input to pyramidal neurons in the aged group. This functional compensation may explain the lack of drastic loss of spontaneous neuronal activity in normal aging.


Subject(s)
Action Potentials/physiology , Aging/pathology , Aging/physiology , Neocortex/ultrastructure , Pyramidal Cells/ultrastructure , Synapses/ultrastructure , Animals , Cell Count , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials/physiology , In Vitro Techniques , Neocortex/physiology , Parietal Lobe/physiology , Parietal Lobe/ultrastructure , Patch-Clamp Techniques , Pyramidal Cells/physiology , Rats , Rats, Inbred BN , Rats, Inbred F344 , Synapses/physiology
20.
J Chem Neuroanat ; 20(1): 71-82, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11074345

ABSTRACT

Deficits in cognitive function have been related to quantitative changes in synaptic population, particularly in the cerebral cortex. Here, we used an established model of perinatal asphyxia that induces morphological changes, i.e. neuron loss in the cerebral cortex and striatum, as well as behavioural deficits. We hypothesized that perinatal asphyxia may lead to a neurodegenerative process resulting in cognitive impairment and altered presynaptic bouton numbers in adult rats. We studied cognitive performance at 18 months and presynaptic bouton numbers at 22 months following perinatal asphyxia. Data of the spatial Morris water escape task did not reveal clear memory or learning deficits in aged asphyctic rats compared to aged control rats. However, a memory impairment in aged rats versus young rats was observed, which was more pronounced in asphyctic rats. We found an increase in presynaptic bouton density in the parietal cortex, whereas no changes were found in striatum and frontal cortex in asphyctic rats. An increase of striatal volume was observed in asphyctic rats, leading to an increase in presynaptic bouton numbers in this area. These findings stress the issue that volume measurements have to be taken into account when determining presynaptic bouton density. Furthermore, perinatal asphyxia led to region-specific changes in presynaptic bouton numbers and it worsened the age-related cognitive impairment. These results suggest that perinatal asphyxia induced neuronal loss, which is compensated for by an increase in presynaptic bouton numbers.


Subject(s)
Asphyxia/pathology , Asphyxia/psychology , Behavior, Animal/physiology , Cerebral Cortex/pathology , Neostriatum/pathology , Presynaptic Terminals/pathology , Adult , Animals , Cognition/physiology , Female , Humans , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Immunohistochemistry , Maze Learning/physiology , Perfusion , Pregnancy , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , Rats, Wistar , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...