Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230234, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-38853565

ABSTRACT

How the two pathognomonic proteins of Alzheimer's disease (AD); amyloid ß (Aß) and tau, cause synaptic failure remains enigmatic. Certain synthetic and recombinant forms of these proteins are known to act concurrently to acutely inhibit long-term potentiation (LTP). Here, we examined the effect of early amyloidosis on the acute disruptive action of synaptotoxic tau prepared from recombinant protein and tau in patient-derived aqueous brain extracts. We also explored the persistence of the inhibition of LTP by different synaptotoxic tau preparations. A single intracerebral injection of aggregates of recombinant human tau that had been prepared by either sonication of fibrils (SτAs) or disulfide bond formation (oTau) rapidly and persistently inhibited LTP in rat hippocampus. The threshold for the acute inhibitory effect of oTau was lowered in amyloid precursor protein (APP)-transgenic rats. A single injection of synaptotoxic tau-containing AD or Pick's disease brain extracts also inhibited LTP, for over two weeks. Remarkably, the persistent disruption of synaptic plasticity by patient-derived brain tau was rapidly reversed by a single intracerebral injection of different anti-tau monoclonal antibodies, including one directed to a specific human tau amino acid sequence. We conclude that patient-derived LTP-disrupting tau species persist in the brain for weeks, maintaining their neuroactivity often in concert with Aß. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Brain , Long-Term Potentiation , tau Proteins , Long-Term Potentiation/drug effects , Animals , tau Proteins/metabolism , Amyloid beta-Peptides/metabolism , Rats , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Brain/metabolism , Rats, Transgenic , Male , Hippocampus/metabolism , Hippocampus/drug effects
2.
Eur J Neurosci ; 58(6): 3402-3411, 2023 09.
Article in English | MEDLINE | ID: mdl-37655756

ABSTRACT

Non-invasive sensory stimulation in the range of the brain's gamma rhythm (30-100 Hz) is emerging as a new potential therapeutic strategy for the treatment of Alzheimer's disease (AD). Here, we investigated the effect of repeated combined exposure to 40 Hz synchronized sound and light stimuli on hippocampal long-term potentiation (LTP) in vivo in three rat models of early AD. We employed a very complete model of AD amyloidosis, amyloid precursor protein (APP)-overexpressing transgenic McGill-R-Thy1-APP rats at an early pre-plaque stage, systemic treatment of transgenic APP rats with corticosterone modelling certain environmental AD risk factors and, importantly, intracerebral injection of highly disease-relevant AD patient-derived synaptotoxic beta-amyloid and tau in wild-type animals. We found that daily treatment with 40 Hz sensory stimulation for 2 weeks fully abrogated the inhibition of LTP in all three models. Moreover, there was a negative correlation between the magnitude of LTP and the level of active caspase-1 in the hippocampus of transgenic APP animals, which suggests that the beneficial effect of 40 Hz stimulation was dependent on modulation of pro-inflammatory mechanisms. Our findings support ongoing clinical trials of gamma-patterned sensory stimulation in early AD.


Subject(s)
Alzheimer Disease , Animals , Rats , Alzheimer Disease/therapy , Neuronal Plasticity , Long-Term Potentiation , Rats, Transgenic , Amyloid beta-Protein Precursor/genetics
3.
J Neurosci ; 43(32): 5870-5879, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37491315

ABSTRACT

Amyloid ß protein (Aß) and tau, the two main proteins implicated in causing Alzheimer's disease (AD), are posited to trigger synaptic dysfunction long before significant synaptic loss occurs in vulnerable circuits. Whereas soluble Aß aggregates from AD brain are well recognized potent synaptotoxins, less is known about the synaptotoxicity of soluble tau from AD or other tauopathy patient brains. Minimally manipulated patient-derived aqueous brain extracts contain the more diffusible native forms of these proteins. Here, we explore how intracerebral injection of Aß and tau present in such aqueous extracts of patient brain contribute to disruption of synaptic plasticity in the CA1 area of the male rat hippocampus. Aqueous extracts of certain AD brains acutely inhibited long-term potentiation (LTP) of synaptic transmission in a manner that required both Aß and tau. Tau-containing aqueous extracts of a brain from a patient with Pick's disease (PiD) also impaired LTP, and diffusible tau from either AD or PiD brain lowered the threshold for AD brain Aß to inhibit LTP. Remarkably, the disruption of LTP persisted for at least 2 weeks after a single injection. These findings support a critical role for diffusible tau in causing rapid onset, persistent synaptic plasticity deficits, and promoting Aß-mediated synaptic dysfunction.SIGNIFICANCE STATEMENT The microtubule-associated protein tau forms relatively insoluble fibrillar deposits in the brains of people with neurodegenerative diseases including Alzheimer's and Pick's diseases. More soluble aggregates of disease-associated tau may diffuse between cells and could cause damage to synapses in vulnerable circuits. We prepared aqueous extracts of diseased cerebral cortex and tested their ability to interfere with synaptic function in the brains of live rats. Tau in these extracts rapidly and persistently disrupted synaptic plasticity and facilitated impairments caused by amyloid ß protein, the other major pathologic protein in Alzheimer's disease. These findings show that certain diffusible forms of tau can mediate synaptic dysfunction and may be a target for therapy.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Male , Rats , Animals , Amyloid beta-Peptides/metabolism , Long-Term Potentiation , Alzheimer Disease/metabolism , tau Proteins/metabolism , Neuronal Plasticity , Synapses/metabolism , Hippocampus/metabolism , Brain/metabolism
4.
Neural Regen Res ; 18(6): 1213-1219, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36453396

ABSTRACT

Cognitive decline in Alzheimer's disease correlates with the extent of tau pathology, in particular tau hyperphosphorylation that initially appears in the transentorhinal and related regions of the brain including the hippocampus. Recent evidence indicates that tau hyperphosphorylation caused by either amyloid-ß or long-term depression, a form of synaptic weakening involved in learning and memory, share similar mechanisms. Studies from our group and others demonstrate that long-term depression-inducing low-frequency stimulation triggers tau phosphorylation at different residues in the hippocampus under different experimental conditions including aging. Conversely, certain forms of long-term depression at hippocampal glutamatergic synapses require endogenous tau, in particular, phosphorylation at residue Ser396. Elucidating the exact mechanisms of interaction between tau and long-term depression may help our understanding of the physiological and pathological functions of tau/tau (hyper)phosphorylation. We first summarize experimental evidence regarding tau-long-term depression interactions, followed by a discussion of possible mechanisms by which this interplay may influence the pathogenesis of Alzheimer's disease. Finally, we conclude with some thoughts and perspectives on future research about these interactions.

5.
J Alzheimers Dis ; 89(1): 335-350, 2022.
Article in English | MEDLINE | ID: mdl-35871344

ABSTRACT

BACKGROUND: Cognitive decline in Alzheimer's disease (AD) correlates with the extent of tau pathology, in particular tau hyperphosphorylation, which is strongly age-associated. Although elevation of cerebrospinal fluid or blood levels of phosphorylated tau (p-Tau) at residues Thr181 (p-Tau181), Thr217 (p-Tau217), and Thr231 (p-Tau231) are proposed to be particularly sensitive markers of preclinical AD, the generation of p-Tau during brain activity is poorly understood. OBJECTIVE: To study whether the expression levels of p-Tau181, p-Tau217, and p-Tau231 can be enhanced by physiological synaptic long-term depression (LTD) which has been linked to the enhancement of p-Tau in hippocampus. METHODS: In vivo electrophysiology was performed in urethane anesthetized young adult and aged male rats. Low frequency electrical stimulation (LFS) was used to induce LTD at CA3 to CA1 synapses. The expression level of p-Tau and total tau was measured in dorsal hippocampus using immunofluorescent staining and/or western blotting. RESULTS: We found that LFS enhanced p-Tau181 and p-Tau217 in an age-dependent manner in the hippocampus of live rats. In contrast, phosphorylation at residues Thr231, Ser202/Thr205, and Ser396 appeared less sensitive to LFS. Pharmacological antagonism of either N-methyl-D-aspartate or metabotropic glutamate 5 receptors inhibited the elevation of both p-Tau181 and p-Tau217. Targeting the integrated stress response, which increases with aging, using a small molecule inhibitor ISRIB, prevented the enhancement of p-Tau by LFS in aged rats. CONCLUSION: Together, our data provide a novel in vivo means to uncover brain plasticity-related cellular and molecular processes of tau phosphorylation at key sites in health and aging.


Subject(s)
Alzheimer Disease , Depression , Alzheimer Disease/cerebrospinal fluid , Animals , Biomarkers/cerebrospinal fluid , Male , Neuronal Plasticity , Phosphorylation , Rats , Synapses/metabolism , tau Proteins/metabolism
6.
Neuropsychopharmacology ; 46(12): 2170-2179, 2021 11.
Article in English | MEDLINE | ID: mdl-34188184

ABSTRACT

Synaptic dysfunction is a likely proximate cause of subtle cognitive impairment in early Alzheimer's disease. Soluble oligomers are the most synaptotoxic forms of amyloid ß-protein (Aß) and mediate synaptic plasticity disruption in Alzheimer's disease amyloidosis. Because the presence and extent of cortisol excess in prodromal Alzheimer's disease predicts the onset of cognitive symptoms we hypothesised that corticosteroids would exacerbate the inhibition of hippocampal synaptic long-term potentiation in a rat model of Alzheimer's disease amyloidosis. In a longitudinal experimental design using freely behaving pre-plaque McGill-R-Thy1-APP male rats, three injections of corticosterone or the glucocorticoid methylprednisolone profoundly disrupted long-term potentiation induced by strong conditioning stimulation for at least 2 months. The same treatments had a transient or no detectible detrimental effect on synaptic plasticity in wild-type littermates. Moreover, corticosterone-mediated cognitive dysfunction, as assessed in a novel object recognition test, was more persistent in the transgenic animals. Evidence for the involvement of pro-inflammatory mechanisms was provided by the ability of the selective the NOD-leucine rich repeat and pyrin containing protein 3 (NLRP3) inflammasome inhibitor Mcc950 to reverse the synaptic plasticity deficit in corticosterone-treated transgenic animals. The marked prolongation of the synaptic plasticity disrupting effects of brief corticosteroid excess substantiates a causal role for hypothalamic-pituitary-adrenal axis dysregulation in early Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloidosis , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/metabolism , Animals , Glucocorticoids , Hypothalamo-Hypophyseal System/metabolism , Male , Neuronal Plasticity , Pituitary-Adrenal System/metabolism , Rats
7.
Front Neurosci ; 13: 861, 2019.
Article in English | MEDLINE | ID: mdl-31474823

ABSTRACT

How endogenously produced soluble amyloid ß-protein (Aß) affects synaptic plasticity in vulnerable circuits should provide insight into early Alzheimer's disease pathophysiology. McGill-R-Thy1-APP transgenic rats, modeling Alzheimer's disease amyloidosis, exhibit an age-dependent soluble Aß-mediated impairment of the induction of long-term potentiation (LTP) by 200 Hz conditioning stimulation at apical CA3-to-CA1 synapses. Here, we investigated if synaptic weakening at these synapses in the form of activity-dependent persistent reversal (depotentiation) of LTP is also altered in pre-plaque rats in vivo. In freely behaving transgenic rats strong, 400 Hz, conditioning stimulation induced stable LTP that was NMDA receptor- and voltage-gated Ca2+ channel-dependent. Surprisingly, the ability of novelty exploration to induce depotentiation of 400 Hz-induced LTP was impaired in an Aß-dependent manner in the freely behaving transgenic rats. Moreover, at apical synapses, low frequency conditioning stimulation (1 Hz) did not trigger depotentiation in anaesthetized transgenic rats, with an age-dependence similar to the LTP deficit. In contrast, at basal synapses neither LTP, induced by 100 or 200 Hz, nor novelty exploration-induced depotentiation was impaired in the freely behaving transgenic rats. These findings indicate that activity-dependent weakening, as well as strengthening, is impaired in a synapse- and age-dependent manner in this model of early Alzheimer's disease amyloidosis.

8.
Neurobiol Dis ; 127: 582-590, 2019 07.
Article in English | MEDLINE | ID: mdl-30910746

ABSTRACT

Soluble synaptotoxic aggregates of the main pathological proteins of Alzheimer's disease, amyloid ß-protein (Aß) and tau, have rapid and potent inhibitory effects on long-term potentiation (LTP). Although the promotion of synaptic weakening mechanisms, including long-term depression (LTD), is posited to mediate LTP inhibition by Aß, little is known regarding the action of exogenous tau on LTD. The present study examined the ability of different assemblies of full-length human tau to affect LTD in the dorsal hippocampus of the anaesthetized rat. Unlike Aß, intracerebroventricular injection of soluble aggregates of tau (SτAs), but not monomers or fibrils, potently increased the threshold for LTD induction in a manner that required cellular prion protein. However, MTEP, an antagonist of the putative prion protein coreceptor metabotropic glutamate receptor 5, did not prevent the disruption of synaptic plasticity by SτAs. In contrast, systemic treatment with Ro 25-6981, a selective antagonist at GluN2B subunit-containing NMDA receptors, reduced SτA-mediated inhibition of LTD, but not LTP. Intriguingly, SτAs completely blocked Aß-facilitated LTD, whereas a subthreshold dose of SτAs facilitated Aß-mediated inhibition of LTP. Overall, these findings support the importance of cellular prion protein in mediating a range of, sometimes opposing, actions of soluble Aß and tau aggregates with different effector mechanisms on synaptic plasticity.


Subject(s)
Amyloid beta-Peptides/pharmacology , Hippocampus/drug effects , Long-Term Synaptic Depression/drug effects , Protein Aggregates/physiology , tau Proteins/metabolism , Animals , Hippocampus/metabolism , Long-Term Synaptic Depression/physiology , Male , Pyridines/pharmacology , Rats , Receptor, Metabotropic Glutamate 5/agonists , Synapses/drug effects , Synapses/physiology , Thiazoles/pharmacology
9.
J Neurosci ; 38(50): 10595-10606, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30355631

ABSTRACT

Intracellular neurofibrillary tangles (NFTs) composed of tau protein are a neuropathological hallmark of several neurodegenerative diseases, the most common of which is Alzheimer's disease (AD). For some time NFTs were considered the primary cause of synaptic dysfunction and neuronal death, however, more recent evidence suggests that soluble aggregates of tau are key drivers of disease. Here we investigated the effect of different tau species on synaptic plasticity in the male rat hippocampus in vivo Intracerebroventricular injection of soluble aggregates formed from either wild-type or P301S human recombinant tau potently inhibited hippocampal long-term potentiation (LTP) at CA3-to-CA1 synapses. In contrast, tau monomers and fibrils appeared inactive. Neither baseline synaptic transmission, paired-pulse facilitation nor burst response during high-frequency conditioning stimulation was affected by the soluble tau aggregates. Similarly, certain AD brain soluble extracts inhibited LTP in a tau-dependent manner that was abrogated by either immunodepletion with, or coinjection of, a mid-region anti-tau monoclonal antibody (mAb), Tau5. Importantly, this tau-mediated block of LTP was prevented by administration of mAbs selective for the prion protein (PrP). Specifically, mAbs to both the mid-region (6D11) and N-terminus (MI-0131) of PrP prevented inhibition of LTP by both recombinant and brain-derived tau. These findings indicate that PrP is a mediator of tau-induced synaptic dysfunction.SIGNIFICANCE STATEMENT Here we report that certain soluble forms of tau selectively disrupt synaptic plasticity in the live rat hippocampus. Further, we show that monoclonal antibodies to cellular prion protein abrogate the impairment of long-term potentiation caused both by recombinant and Alzheimer's disease brain-derived soluble tau. These findings support a critical role for cellular prion protein in the deleterious synaptic actions of extracellular soluble tau in tauopathies, including Alzheimer's disease. Thus, approaches targeting cellular prion protein, or downstream pathways, might provide an effective strategy for developing therapeutics.


Subject(s)
Hippocampus/metabolism , Hippocampus/pathology , Neuronal Plasticity/physiology , PrPC Proteins/metabolism , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Angiogenesis Inhibitors/pharmacology , Animals , Female , Hippocampus/drug effects , Humans , Male , Neuronal Plasticity/drug effects , Prion Proteins/metabolism , Rats
10.
Neuropharmacology ; 121: 231-246, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28390893

ABSTRACT

Alzheimer's disease amyloid-ß (Aß) oligomers are synaptotoxic, inappropriately increasing extracellular glutamate concentration and glutamate receptor activation to thereby rapidly disrupt synaptic plasticity. Thus, acutely promoting brain glutamate homeostasis with a blood-based scavenging system, glutamate-oxaloacetate transaminase (GOT), and blocking metabotropic glutamate 5 (mGlu5) receptor or its co-receptor cellular prion protein (PrP), prevent the acute inhibition of long-term potentiation (LTP) by exogenous Aß. Here, we evaluated the time course of the effects of such interventions in the persistent disruptive effects of Aß oligomers, either exogenously injected in wild type rats or endogenously generated in transgenic rats that model Alzheimer's disease amyloidosis. We report that repeated, but not acute, systemic administration of recombinant GOT type 1, with or without the glutamate co-substrate oxaloacetate, reversed the persistent deleterious effect of exogenous Aß on synaptic plasticity. Moreover, similar repetitive treatment reversibly abrogated the inhibition of LTP monitored longitudinally in freely behaving transgenic rats. Remarkably, brief repeated treatment with an mGlu5 receptor antagonist, basimglurant, or an antibody that prevents Aß oligomer binding to PrP, ICSM35, also had similar reversible ameliorative effects in the transgenic rat model. Overall, the present findings support the ongoing development of therapeutics for early Alzheimer's disease based on these complementary approaches.


Subject(s)
Amyloidosis/pathology , Amyloidosis/physiopathology , CA1 Region, Hippocampal/physiology , Long-Term Potentiation/drug effects , Prion Proteins/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/blood , Amyloidosis/genetics , Animals , Antibodies/pharmacology , Aspartate Aminotransferase, Cytoplasmic/pharmacology , CA1 Region, Hippocampal/drug effects , Disease Models, Animal , Humans , Imidazoles/pharmacology , Long-Term Potentiation/genetics , Male , Mutation/genetics , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Protein Binding/drug effects , Protein Binding/genetics , Pyridines/pharmacology , Rats , Rats, Transgenic , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Wakefulness
11.
Hippocampus ; 26(12): 1655-1665, 2016 12.
Article in English | MEDLINE | ID: mdl-27701797

ABSTRACT

Aggregated amyloid ß-protein (Aß) is pathognomonic of Alzheimer's disease and certain assemblies of Aß are synaptotoxic. Excess glutamate or diminished glutathione reserve are both implicated in mediating or modulating Aß-induced disruption of synaptic plasticity. The system xc- antiporter promotes Na+ -independent exchange of cystine with glutamate thereby providing a major source of extracellular glutamate and intracellular glutathione concentrations. Here we probed the ability of two drugs with opposite effects on system xc-, the inhibitor sulfasalazine and facilitator N-acetylcysteine, to modulate the ability of Aß1-42 to inhibit long-term potentiation (LTP) in the CA1 area of the anaesthetized rat. Whereas acute systemic treatment with sulfasalazine lowered the threshold for Aß to interfere with synaptic plasticity, N-acetylcysteine prevented the inhibition of LTP by Aß alone or in combination with sulfasalazine. Moreover acute N-acetylcysteine also prevented the inhibition of LTP by TNFα, a putative mediator of Aß actions, and repeated systemic N-acetylcysteine treatment for 7 days reversed the delayed deleterious effect of Aß on LTP. Since both of these drugs are widely used clinically, further evaluation of their potential beneficial and deleterious actions in early Alzheimer's disease seems warranted. © 2016 Wiley Periodicals, Inc.


Subject(s)
Acetylcysteine/pharmacology , Amyloid beta-Peptides/toxicity , Central Nervous System Agents/pharmacology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Peptide Fragments/toxicity , Sulfasalazine/pharmacology , Amino Acid Transport Systems, Acidic/antagonists & inhibitors , Amino Acid Transport Systems, Acidic/genetics , Amino Acid Transport Systems, Acidic/metabolism , Animals , Catheters, Indwelling , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glutathione/metabolism , Hippocampus/metabolism , Long-Term Potentiation/physiology , Male , Maleates/pharmacology , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
12.
J Neurosci ; 35(16): 6265-76, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904780

ABSTRACT

Alzheimer's disease (AD) and familial Danish dementia (FDD) are degenerative neurological diseases characterized by amyloid pathology. Normal human sera contain IgG antibodies that specifically bind diverse preamyloid and amyloid proteins and have shown therapeutic potential in vitro and in vivo. We cloned one of these antibodies, 3H3, from memory B cells of a healthy individual using a hybridoma method. 3H3 is an affinity-matured IgG that binds a pan-amyloid epitope, recognizing both Aß and λ Ig light chain (LC) amyloids, which are associated with AD and primary amyloidosis, respectively. The pan-amyloid-binding properties of 3H3 were demonstrated using ELISA, immunohistochemical studies, and competition binding assays. Functional studies showed that 3H3 inhibits both Aß and LC amyloid formation in vitro and abrogates disruption of hippocampal synaptic plasticity by AD-patient-derived soluble Aß in vivo. A 3H3 single-chain variable fragment (scFv) retained the binding specificity of the 3H3 IgG and, when expressed in the brains of transgenic mice using an adeno-associated virus (AAV) vector, decreased parenchymal Aß amyloid deposition in TgCRND8 mice and ADan (Danish Amyloid) cerebral amyloid angiopathy in the mouse model of FDD. These data indicate that naturally occurring human IgGs can recognize a conformational, amyloid-specific epitope and have potent anti-amyloid activities, providing a rationale to test their potential as antibody therapeutics for diverse neurological and other amyloid diseases.


Subject(s)
Amyloid beta-Peptides/immunology , Amyloid/metabolism , Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Amyloid/drug effects , Animals , Antibodies, Monoclonal/pharmacology , Brain/metabolism , Cataract/immunology , Cerebellar Ataxia/immunology , Cerebral Amyloid Angiopathy/immunology , Deafness/immunology , Dementia/immunology , Humans , Immunoglobulin G/pharmacology , Male , Mice , Mice, Transgenic , Rats
13.
Philos Trans R Soc Lond B Biol Sci ; 369(1633): 20130147, 2014 Jan 05.
Article in English | MEDLINE | ID: mdl-24298149

ABSTRACT

Many endogenous factors influence the time course and extent of the detrimental effects of amyloid ß-protein (Aß) on synaptic function. Here, we assessed the impact of varying endogenous glutamatergic and cholinergic transmission by pharmacological means on the disruption of plasticity at hippocampal CA3-to-CA1 synapses in the anaesthetized rat. NMDA receptors (NMDARs) are considered critical in mediating Aß-induced inhibition of long-term potentiation (LTP). However, intracerebroventricular injection of Aß1-42 inhibited not only NMDAR-dependent LTP but also voltage-activated Ca(2+)-dependent LTP induced by strong conditioning stimulation during NMDAR blockade. On the other hand, another form of NMDAR-independent synaptic plasticity, endogenous acetylcholine-induced muscarinic receptor-dependent long-term enhancement, was not hindered by Aß1-42. Interestingly, augmenting endogenous acetylcholine activation of nicotinic receptors prior to the injection of Aß1-42 prevented the inhibition of NMDAR-dependent LTP, whereas the same intervention when introduced after the infusion of Aß was ineffective. We also examined the duration of action of Aß, including water soluble Aß from Alzheimer's disease (AD) brain. Remarkably, the inhibition of LTP induction caused by a single injection of sodium dodecyl sulfate-stable Aß dimer-containing AD brain extract persisted for at least a week. These findings highlight the need to increase our understanding of non-NMDAR mechanisms and of developing novel means of overcoming, rather than just preventing, the deleterious synaptic actions of Aß.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Peptide Fragments/metabolism , Synapses/physiology , Acetylcholine/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Analysis of Variance , Animals , Brain Chemistry , Excitatory Postsynaptic Potentials/physiology , Extracellular Fluid/metabolism , Humans , Long-Term Potentiation/drug effects , Male , Peptide Fragments/pharmacology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Nicotinic/metabolism , Synapses/drug effects
14.
Pharmacol Biochem Behav ; 100(4): 855-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21536064

ABSTRACT

The cognitive and related symptoms of Alzheimer's disease are mainly attributable to synaptic failure. Here we review recent research on how the Alzheimer's disease amyloid ß-protein (Aß) affects glutamate receptors and fast excitatory synaptic transmission and plasticity of that transmission. l-glutamate, the main excitatory neurotransmitter in the brain, has long been implicated in causing NMDA receptor-mediated excitotoxicity leading to neurodegeneration in the late stages of the disease. However there is now extensive evidence that soluble Aß oligomers disrupt synaptic transmission and especially synaptic plasticity via non-excitotoxic glutamatergic mechanisms. New data highlight the relatively selective involvement of certain glutamate receptor subtypes including GluN2B (NR2B) subunit-containing NMDA receptors and mGlu5 receptors. Aß exerts direct and indirect effects on synaptic plasticity-related glutamate receptor signaling and trafficking between different neuronal compartments. For example, Aß-induced ectopic NMDA and mGlu receptor-mediated signaling coupled with caspase-3 activation may cause inhibition of long-term potentiation and facilitation of long-term depression. Intriguingly, some of the disruptive synaptic actions of Aß have been found to be dependent on endogenous tau located in dendrites or spines. Given the role of glutamatergic transmission in regulating Aß production and release, future therapies targeting glutamate offer the opportunity to remedy both mis-processing of Aß and cellular mechanisms of synaptic failure in early AD.


Subject(s)
Alzheimer Disease/physiopathology , Receptors, Glutamate/physiology , Humans , Signal Transduction
15.
Eur J Pharmacol ; 677(1-3): 63-70, 2012 Feb 29.
Article in English | MEDLINE | ID: mdl-22200627

ABSTRACT

Nicotinic acetylcholine receptors mediate fast cholinergic modulation of glutamatergic transmission and synaptic plasticity. Here we investigated the effects of subtype selective activation of the α7 nicotinic acetylcholine receptors on hippocampal transmission and the inhibition of synaptic long-term potentiation by the Alzheimer's disease associated amyloid ß-protein (Aß). The α7 nicotinic acetylcholine receptor agonist "compound A" ((R)-N-(1-azabicyclo[2.2.2]oct-3-yl)(5-(2-pyridyl))thiophene-2-carboxamide) induced a rapid-onset persistent enhancement of synaptic transmission in the dentate gyrus in vitro. Consistent with a requirement for activation of α7 nicotinic acetylcholine receptors, the type II α7-selective positive allosteric modulator PheTQS ((3aR, 4S, 9bS)-4-(4-methylphenyl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinoline-8-sulfonamide) potentiated, and the antagonist methyllycaconitine (MLA) prevented the persistent enhancement. Systemic injection of the agonist also induced a similar MLA-sensitive persistent enhancement of synaptic transmission in the CA1 area in vivo. Remarkably, although compound A did not affect control long-term potentiation (LTP) in vitro, it prevented the inhibition of LTP by Aß1-42 and this effect was inhibited by MLA. These findings strongly indicate that activation of α7 nicotinic acetylcholine receptors is sufficient to persistently enhance hippocampal synaptic transmission and to overcome the inhibition of LTP by Aß.


Subject(s)
Amyloid beta-Peptides/pharmacology , Hippocampus/cytology , Hippocampus/physiology , Long-Term Potentiation/drug effects , Peptide Fragments/pharmacology , Receptors, Nicotinic/metabolism , Synaptic Transmission/drug effects , Allosteric Regulation/drug effects , Animals , Drug Interactions , Hippocampus/drug effects , In Vitro Techniques , Male , Nicotinic Agonists/pharmacology , Rats , Rats, Wistar , Sulfonamides/chemistry , Sulfonamides/pharmacology , Thiophenes/chemistry , Thiophenes/pharmacology , alpha7 Nicotinic Acetylcholine Receptor
16.
J Neurosci ; 31(11): 3953-61, 2011 Mar 16.
Article in English | MEDLINE | ID: mdl-21411638

ABSTRACT

There is accumulating evidence that sleep contributes to memory formation and learning, but the underlying cellular mechanisms are incompletely understood. To investigate the impact of sleep on excitatory synaptic transmission, we obtained whole-cell patch-clamp recordings from layer V pyramidal neurons in acute slices of somatosensory cortex of juvenile rats (postnatal days 21-25). In animals after the dark period, philanthotoxin 74 (PhTx)-sensitive calcium-permeable AMPA receptors (CP-AMPARs) accounted for ∼25% of total EPSP size, and current-voltage (I-V) relationships of the underlying EPSCs showed inward rectification. In contrast, in similar experiments after the light period, EPSPs were PhTx insensitive with linear I-V characteristics, indicating that CP-AMPARs were less abundant. Combined EEG and EMG recordings confirmed that slow-wave sleep-associated delta wave power peaked at the onset of the more quiescent, lights-on phase of the cycle. Subsequently, we show that burst firing, a characteristic action potential discharge mode of layer V pyramidal neurons during slow-wave sleep has a dual impact on synaptic AMPA receptor composition: repetitive burst firing without synaptic stimulation eliminated CP-AMPARs by activating serine/threonine phosphatases. Additionally, repetitive burst-firing paired with EPSPs led to input-specific long-term depression (LTD), affecting Ca(2+) impermeable AMPARs via protein kinase C signaling. In agreement with two parallel mechanisms, simple bursts were ineffective after the light period but paired bursts induced robust LTD. In contrast, incremental LTD was generated by both conditioning protocols after the dark cycle. Together, our results demonstrate qualitative changes at neocortical glutamatergic synapses that can be induced by discharge patterns characteristic of non-rapid eye movement sleep.


Subject(s)
Calcium/metabolism , Neurons/physiology , Receptors, AMPA/physiology , Sleep/physiology , Somatosensory Cortex/physiology , Action Potentials/drug effects , Action Potentials/physiology , Analysis of Variance , Animals , Electroencephalography , Electromyography , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Neurons/drug effects , Phenols/pharmacology , Polyamines/pharmacology , Rats , Rats, Wistar , Synapses/drug effects , Synapses/physiology
17.
Neuromolecular Med ; 12(1): 13-26, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19757208

ABSTRACT

Alzheimer's disease (AD) is characterized neuropathologically by the deposition of different forms of amyloid beta-protein (A beta) including variable amounts of soluble species that correlate with severity of dementia. The extent of synaptic loss in the brain provides the best morphological correlate of cognitive impairment in clinical AD. Animal research on the pathophysiology of AD has therefore focussed on how soluble A beta disrupts synaptic mechanisms in vulnerable brain regions such as the hippocampus. Synaptic plasticity in the form of persistent activity-dependent increases or decreases in synaptic strength provide a neurophysiological substrate for hippocampal-dependent learning and memory. Acute treatment with human-derived or chemically prepared soluble A beta that contains certain oligomeric assemblies, potently and selectively disrupts synaptic plasticity causing inhibition of long-term potentiation (LTP) and enhancement of long-term depression (LTD) of glutamatergic transmission. Over time these and related actions of A beta have been implicated in reducing synaptic integrity. This review addresses the involvement of neurotransmitter intercellular signaling in mediating or modulating the synaptic plasticity disrupting actions of soluble A beta, with particular emphasis on the different roles of glutamatergic and cholinergic mechanisms. There is growing evidence to support the view that NMDA and possibly nicotinic receptors are critically involved in mediating the disruptive effect of A beta and that targeting muscarinic receptors can indirectly modulate A beta's actions. Such studies should help inform ongoing and future clinical trials of drugs acting through the glutamatergic and cholinergic systems.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Acetylcholine/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Glutamic Acid/metabolism , Humans , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Receptors, Cholinergic/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
18.
Cell Mol Neurobiol ; 26(7-8): 1521-39, 2006.
Article in English | MEDLINE | ID: mdl-16691438

ABSTRACT

1. After traumatic spinal cord injury (SCI), histological and neurological consequences are developing for several days and even weeks. However, little is known about the dynamics of changes in spinal axonal conductivity. The aim of this study was to record and compare repeated spinal cord evoked potentials (SCEP) after SCI in the rat during a 4 weeks' interval. These recordings were used: (i) for studying the dynamics of functional changes in spinal axons after SCI, and (ii) to define the value of SCEP as an independent outcome parameter in SCI studies. 2. We have used two pairs of chronically implanted epidural electrodes for stimulation/recording. The electrodes were placed below and above the site of injury, respectively. Animals with implanted electrodes underwent spinal cord compression injury induced by epidural balloon inflation at Th8-Th9 level. There were five experimental groups of animals, including one control group (sham-operated, no injury), and four injury groups (different degrees of SCI). 3. After SCI, SCEP waveform was either significantly reduced or completely lost. Partial recovery of SCEPs was observed in all groups. The onset and extent of recovery clearly correlated with the severity of injury. There was good correlation between quantitated SCEP variables and the volumes of the compressing balloon. However, sensitivity of electropohysiological parameters was inferior compared to neurological and morphometric outcomes. 4. Our study shows for the first time, that the dynamics of axonal recovery depends on the degree of injury. After mild injury, recovery of signal is rapid. However, after severe injury, axonal conductivity can re-appear after as long as 2 weeks postinjury. In conclusion, SCEPs can be used as an independent parameter of outcome after SCI, but in general, the sensitivity of electrophysiological data were worse than standard morphological and neurological evaluations.


Subject(s)
Evoked Potentials , Spinal Cord Injuries/physiopathology , Spinal Cord/physiology , Animals , Body Weight , Electric Stimulation , Electrophysiology , Locomotion , Male , Rats , Rats, Wistar , Spinal Cord Injuries/mortality , Spinal Cord Injuries/rehabilitation , Time
19.
J Neurosci Methods ; 141(1): 125-34, 2005 Jan 30.
Article in English | MEDLINE | ID: mdl-15585296

ABSTRACT

We have recorded and characterized the spinal cord evoked potentials (SCEPs) from the epidural space in the halothane-anesthetized rats. A group of 11 adult Wistar male rats was chronically implanted with two pairs of epidural electrodes. SCEPs were repeatedly elicited by applying electrical stimuli via bipolar U-shaped electrodes to the dorsal aspect of the spinal cord at C3-4 or Th11-12 levels, respectively. Responses were registered with the other pair of implanted electrodes, thus allowing us to monitor the descending (stimulation cervical/recording thoracic) and ascending SCEPs (stimulation thoracic/recording cervical). We studied the time-dependent changes of several SCEP parameters, among them the latency and amplitude of two major negative waves N1 and N2. During 4-weeks' survival, all major components of recordings remained stable and only minor changes in some parameters of the SCEPs were detected. We concluded that this technique enables repeated quantitative analysis of the conductivity of the spinal cord white matter in the rat. Our results indicate that SCEPs could be used in long-term experiments for monitoring progressive changes (degeneration/regeneration) in long projection tracts, primarily those occupying the dorsolateral quadrants of the spinal cord. These include projections that are of interest in spinal cord injury studies, i.e. ascending primary afferents, and important descending pathways including corticospinal, rubrospinal, reticulospinal, raphespinal and vestibulospinal tracts.


Subject(s)
Action Potentials/physiology , Axons/physiology , Electrophysiology/instrumentation , Neural Pathways/physiology , Neurophysiology/instrumentation , Spinal Cord/physiology , Animals , Electrodes, Implanted , Electrophysiology/methods , Epidural Space/surgery , Evoked Potentials/physiology , Male , Neural Conduction/physiology , Neural Pathways/anatomy & histology , Neurophysiology/methods , Neurosurgical Procedures/methods , Rats , Rats, Wistar , Reaction Time/physiology , Spinal Cord/anatomy & histology , Spinal Cord/surgery , Time Factors
20.
Cell Mol Neurobiol ; 24(3): 477-89, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15206826

ABSTRACT

1. To test our hypothesis that a transient nonlethal ischemic insult benefits the lumbosacral spinal cord ischemic injury, nestin, the marker of proliferating cells, and Fluoro-Jade B, the marker of degenerating cells, were used in rats. Morphological outcome was evaluated after 12-min ischemia versus 12-min ischemia preconditioned by 3-min ischemic period and 30-min recirculation (IPC), in each group followed by 2, 3, and 4 days of posttreatment survival. 2. Twelve-minute ischemia, inducing nestin-positivity in ependyma and reactive astrocytes at the L(1-3) spinal cord segments, shows this region as the viable region of spinal cord in all postischemic survival periods. On the other hand, abundance of Fluoro-Jade B-positive cells, distributed throughout the dorsal horn and intermediate zone of L4-S2 segments, points out the most injured spinal cord region by ischemia. 3. After the same ischemic insult in IPC rats only a few nestin-positive ependymal cell and reactive astrocytes appeared beside the nestin-positive vessels in the lower lumbar and sacral spinal cord segments of all survival periods. The appearance of nestin-positive cells in the spinal cord segments, which "should have been affected" by ischemia indicates protection of this region by the IPC treatment. 4. The number and density evaluation of Fluoro-Jade B fluorescent cells of L4-S2 segments after ischemia and IPC confirmed that degenerating cells were significantly reduced in the IPC rats in all survival periods. 5. Our results showing the immunohistochemical response of epemdyma, committed to the presence of viable tissue, indicate that the ependymal cells may contribute to the ischemic resistance in the IPC rats.


Subject(s)
Ependyma/metabolism , Ischemic Preconditioning , Nerve Degeneration/metabolism , Nerve Regeneration/physiology , Spinal Cord Ischemia/metabolism , Spinal Cord/metabolism , Animals , Astrocytes/metabolism , Biomarkers , Disease Models, Animal , Ependyma/cytology , Fluoresceins , Fluorescent Dyes , Gliosis/metabolism , Gliosis/physiopathology , Immunohistochemistry , Intermediate Filament Proteins/biosynthesis , Lumbar Vertebrae , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Nerve Tissue Proteins/biosynthesis , Nestin , Organic Chemicals , Rats , Rats, Wistar , Spinal Cord/cytology , Spinal Cord/physiopathology , Spinal Cord Ischemia/physiopathology , Spinal Cord Ischemia/prevention & control , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...