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










Publication year range
1.
Brain Behav Immun ; 119: 286-300, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38608739

ABSTRACT

Alzheimer's disease is a progressive neurological disorder causing memory loss and cognitive decline. The underlying causes of cognitive deterioration and neurodegeneration remain unclear, leading to a lack of effective strategies to prevent dementia. Recent evidence highlights the role of neuroinflammation, particularly involving microglia, in Alzheimer's disease onset and progression. Characterizing the initial phase of Alzheimer's disease can lead to the discovery of new biomarkers and therapeutic targets, facilitating timely interventions for effective treatments. We used the AppNL-G-F knock-in mouse model, which resembles the amyloid pathology and neuroinflammatory characteristics of Alzheimer's disease, to investigate the transition from a pre-plaque to an early plaque stage with a combined functional and molecular approach. Our experiments show a progressive decrease in the power of cognition-relevant hippocampal gamma oscillations during the early stage of amyloid pathology, together with a modification of fast-spiking interneuron intrinsic properties and postsynaptic input. Consistently, transcriptomic analyses revealed that these effects are accompanied by changes in synaptic function-associated pathways. Concurrently, homeostasis- and inflammatory-related microglia signature genes were downregulated. Moreover, we found a decrease in Iba1-positive microglia in the hippocampus that correlates with plaque aggregation and neuronal dysfunction. Collectively, these findings support the hypothesis that microglia play a protective role during the early stages of amyloid pathology by preventing plaque aggregation, supporting neuronal homeostasis, and overall preserving the oscillatory network's functionality. These results suggest that the early alteration of microglia dynamics could be a pivotal event in the progression of Alzheimer's disease, potentially triggering plaque deposition, impairment of fast-spiking interneurons, and the breakdown of the oscillatory circuitry in the hippocampus.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Disease Progression , Hippocampus , Mice, Transgenic , Microglia , Plaque, Amyloid , Animals , Microglia/metabolism , Microglia/pathology , Hippocampus/metabolism , Hippocampus/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Amyloid beta-Peptides/metabolism , Male , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Interneurons/metabolism , Interneurons/pathology
2.
Front Cell Neurosci ; 18: 1347535, 2024.
Article in English | MEDLINE | ID: mdl-38650656

ABSTRACT

Astrocytes represent the most abundant cell type in the brain, where they play critical roles in synaptic transmission, cognition, and behavior. Recent discoveries show astrocytes are involved in synaptic dysfunction during Alzheimer's disease (AD). AD patients have imbalanced cholesterol metabolism, demonstrated by high levels of side-chain oxidized cholesterol known as 27-hydroxycholesterol (27-OH). Evidence from our laboratory has shown that elevated 27-OH can abolish synaptic connectivity during neuromaturation, but its effect on astrocyte function is currently unclear. Our results suggest that elevated 27-OH decreases the astrocyte function in vivo in Cyp27Tg, a mouse model of brain oxysterol imbalance. Here, we report a downregulation of glutamate transporters in the hippocampus of CYP27Tg mice together with increased GFAP. GLT-1 downregulation was also observed when WT mice were fed with high-cholesterol diets. To study the relationship between astrocytes and neurons, we have developed a 3D co-culture system that allows all the cell types from mice embryos to differentiate in vitro. We report that our 3D co-cultures reproduce the effects of 27-OH observed in 2D neurons and in vivo. Moreover, we found novel degenerative effects in astrocytes that do not appear in 2D cultures, together with the downregulation of glutamate transporters GLT-1 and GLAST. We propose that this transporter dysregulation leads to neuronal hyperexcitability and synaptic dysfunction based on the effects of 27-OH on astrocytes. Taken together, these results report a new mechanism linking oxysterol imbalance in the brain and synaptic dysfunction through effects on astrocyte function.

3.
Prog Neurobiol ; 235: 102585, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38367747

ABSTRACT

Alzheimer's disease (AD) is a multifactorial disorder driven by abnormal amyloid ß-peptide (Aß) levels. In this study, we investigated the role of presenilin-like signal peptide peptidase-like 2b (SPPL2b) in AD pathophysiology and its potential as a druggable target within the Aß cascade. Exogenous Aß42 influenced SPPL2b expression in human cell lines and acute mouse brain slices. SPPL2b and its AD-related substrate BRI2 were evaluated in the brains of AppNL-G-F knock-in AD mice and human postmortem AD brains. An early high cortical expression of SPPL2b was observed, followed by a downregulation in late AD pathology in AppNL-G-F mice, correlating with synaptic loss. To understand the consequences of pathophysiological SPPL2b dysregulation, we found that SPPL2b overexpression significantly increased APP cleavage, while genetic deletion reduced APP cleavage and Aß production. Notably, postmortem AD brains showed higher levels of SPPL2b's BRI2 substrate compared to healthy control samples. These results strongly support the involvement of SPPL2b in AD pathology. The early Aß-induced upregulation of SPPL2b may enhance Aß production in a vicious cycle, further aggravating Aß pathology. Therefore, SPPL2b emerges as a potential anti-Aß drug target.


Subject(s)
Alzheimer Disease , Animals , Humans , Mice , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Disease Models, Animal
4.
Exp Neurol ; 369: 114545, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37726047

ABSTRACT

Alzheimer's disease (AD) is characterized at an early stage by memory alterations that worsen during the development of the disease. Several clinical trials in phase 3 have failed despite being able to counteract classical AD-related alterations, possibly because of the lack of recovery of the regular neuronal network activity essential for memory including low gamma oscillations (γ-Osc). Nowadays, Levetiracetam (LEV), an SV2A modulator approved for epilepsy, is being used in trials with AD patients without further support for neurophysiological relevant effects on restoring the normal function of hippocampal neuronal network activity. Using concomitant recordings of local field potential γ-Osc and patch-clamp recordings of fast-spiking interneurons (FS-IN) on hippocampal slices of WT and AppNL-G-F AD animals, we found that LEV restores the power and rhythmicity of γ-Osc previously reduced by acute application of amyloid-ß on WT hippocampal slices, this effect is accompanied by the recovery of the synchronicity in the firing of FS-IN. In addition, we found that LEV counteracts the hippocampal γ-Osc alterations in the early prodromal stage of the disease in AppNL-G-F mice by recovering the rhythmicity of γ-Osc and the synchronicity in the firing of FS-IN. Altogether the results show that the precise modulation of neuronal circuits with LEV is a promising strategy to counteract early-stage alterations in hippocampal activity by modulating FS-IN in a memory-relevant neuronal network state like γ-Osc.

5.
ACS Chem Neurosci ; 2023 Apr 06.
Article in English | MEDLINE | ID: mdl-37023330

ABSTRACT

Self-replication of amyloid-ß-peptide (Aß) fibril formation is a hallmark in Alzheimer's disease (AD). Detailed insights have been obtained in Aß self-assembly in vitro, yet whether similar mechanisms are relevant in vivo has remained elusive. Here, we investigated the ability of in vivo-derived Aß fibrils from two different amyloid precursor protein knock-in AD mouse models to seed Aß42 aggregation, where we quantified the microscopic rate constants. We found that the nucleation mechanism of in vivo-derived fibril-seeded Aß42 aggregation can be described with the same kinetic model as that in vitro. Further, we identified the inhibitory mechanism of the anti-amyloid BRICHOS chaperone on seeded Aß42 fibrillization, revealing a suppression of secondary nucleation and fibril elongation, which is strikingly similar as observed in vitro. These findings hence provide a molecular understanding of the Aß42 nucleation process triggered by in vivo-derived Aß42 propagons, providing a framework for the search for new AD therapeutics.

6.
Mol Psychiatry ; 28(6): 2177-2188, 2023 06.
Article in English | MEDLINE | ID: mdl-36991134

ABSTRACT

In the mammalian brain information processing and storage rely on the complex coding and decoding events performed by neuronal networks. These actions are based on the computational ability of neurons and their functional engagement in neuronal assemblies where precise timing of action potential firing is crucial. Neuronal circuits manage a myriad of spatially and temporally overlapping inputs to compute specific outputs that are proposed to underly memory traces formation, sensory perception, and cognitive behaviors. Spike-timing-dependent plasticity (STDP) and electrical brain rhythms are suggested to underlie such functions while the physiological evidence of assembly structures and mechanisms driving both processes continues to be scarce. Here, we review foundational and current evidence on timing precision and cooperative neuronal electrical activity driving STDP and brain rhythms, their interactions, and the emerging role of glial cells in such processes. We also provide an overview of their cognitive correlates and discuss current limitations and controversies, future perspectives on experimental approaches, and their application in humans.


Subject(s)
Brain , Neurons , Animals , Humans , Brain/physiology , Neurons/physiology , Neuronal Plasticity/physiology , Periodicity , Action Potentials/physiology , Neuroglia , Models, Neurological , Mammals
7.
Transl Neurodegener ; 12(1): 6, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36740709

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a progressive multifaceted neurodegenerative disorder for which no disease-modifying treatment exists. Neuroinflammation is central to the pathology progression, with evidence suggesting that microglia-released galectin-3 (gal3) plays a pivotal role by amplifying neuroinflammation in AD. However, the possible involvement of gal3 in the disruption of neuronal network oscillations typical of AD remains unknown. METHODS: Here, we investigated the functional implications of gal3 signaling on experimentally induced gamma oscillations ex vivo (20-80 Hz) by performing electrophysiological recordings in the hippocampal CA3 area of wild-type (WT) mice and of the 5×FAD mouse model of AD. In addition, the recorded slices from WT mice under acute gal3 application were analyzed with RT-qPCR to detect expression of some neuroinflammation-related genes, and amyloid-ß (Aß) plaque load was quantified by immunostaining in the CA3 area of 6-month-old 5×FAD mice with or without Gal3 knockout (KO). RESULTS: Gal3 application decreased gamma oscillation power and rhythmicity in an activity-dependent manner, which was accompanied by impairment of cellular dynamics in fast-spiking interneurons (FSNs) and pyramidal cells. We found that the gal3-induced disruption was mediated by the gal3 carbohydrate-recognition domain and prevented by the gal3 inhibitor TD139, which also prevented Aß42-induced degradation of gamma oscillations. Furthermore, the 5×FAD mice lacking gal3 (5×FAD-Gal3KO) exhibited WT-like gamma network dynamics and decreased Aß plaque load. CONCLUSIONS: We report for the first time that gal3 impairs neuronal network dynamics by spike-phase uncoupling of FSNs, inducing a network performance collapse. Moreover, our findings suggest gal3 inhibition as a potential therapeutic strategy to counteract the neuronal network instability typical of AD and other neurological disorders encompassing neuroinflammation and cognitive decline.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Galectin 3/genetics , Galectin 3/therapeutic use , Mice, Transgenic , Neuroinflammatory Diseases , Amyloid beta-Peptides/metabolism , Interneurons/metabolism , Interneurons/pathology , Plaque, Amyloid
8.
RSC Chem Biol ; 3(11): 1342-1358, 2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36349220

ABSTRACT

Proteins can self-assemble into amyloid fibrils or amorphous aggregates and thereby cause disease. Molecular chaperones can prevent both these types of protein aggregation, but to what extent the respective mechanisms are overlapping is not fully understood. The BRICHOS domain constitutes a disease-associated chaperone family, with activities against amyloid neurotoxicity, fibril formation, and amorphous protein aggregation. Here, we show that the activities of BRICHOS against amyloid-induced neurotoxicity and fibril formation, respectively, are oppositely dependent on a conserved aspartate residue, while the ability to suppress amorphous protein aggregation is unchanged by Asp to Asn mutations. The Asp is evolutionarily highly conserved in >3000 analysed BRICHOS domains but is replaced by Asn in some BRICHOS families. The conserved Asp in its ionized state promotes structural flexibility and has a pK a value between pH 6.0 and 7.0, suggesting that chaperone effects can be differently affected by physiological pH variations.

9.
Prog Neurobiol ; 219: 102366, 2022 12.
Article in English | MEDLINE | ID: mdl-36273719

ABSTRACT

The pro-inflammatory and highly amyloidogenic protein S100A9 is central to the amyloid-neuroinflammatory cascade in neurodegenerative diseases leading to cognitive impairment. Molecular chaperone activity of Bri2 BRICHOS has been demonstrated against a range of amyloidogenic polypeptides. Using a combination of thioflavin T fluorescence kinetic assay, atomic force microscopy and immuno electron microscopy we show here that recombinant Bri2 BRICHOS effectively inhibits S100A9 amyloid growth by capping amyloid fibrils. Using ex-vivo neuronal network electrophysiology in mouse brain slices we also show that both native S100A9 and amyloids of S100A9 disrupt cognition-relevant gamma oscillation power and rhythmicity in hippocampal area CA3 in a time- and protein conformation-dependent manner. Both effects were associated with Toll-like receptor 4 (TLR4) activation and were not observed upon TLR4 blockade. Importantly, S100A9 that had co-aggregated with Bri2 BRICHOS did not elicit degradation of gamma oscillations. Taken together, this work provides insights on the potential influence of S100A9 on cognitive dysfunction in Alzheimer's disease (AD) via gamma oscillation impairment from experimentally-induced gamma oscillations, and further highlights Bri2 BRICHOS as a chaperone against detrimental effects of amyloid self-assembly.


Subject(s)
Alzheimer Disease , Toll-Like Receptor 4 , Animals , Mice , Alzheimer Disease/metabolism , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Amyloidogenic Proteins/metabolism , Calgranulin B/metabolism , Toll-Like Receptor 4/metabolism , CA3 Region, Hippocampal/metabolism
10.
Commun Biol ; 5(1): 245, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35314851

ABSTRACT

Sustained microglial activation and increased pro-inflammatory signalling cause chronic inflammation and neuronal damage in Alzheimer's disease (AD). Resolution of inflammation follows neutralization of pathogens and is a response to limit damage and promote healing, mediated by pro-resolving lipid mediators (LMs). Since resolution is impaired in AD brains, we decided to test if intranasal administration of pro-resolving LMs in the AppNL-G-F/NL-G-F mouse model for AD could resolve inflammation and ameliorate pathology in the brain. A mixture of the pro-resolving LMs resolvin (Rv) E1, RvD1, RvD2, maresin 1 (MaR1) and neuroprotectin D1 (NPD1) was administered to stimulate their respective receptors. We examined amyloid load, cognition, neuronal network oscillations, glial activation and inflammatory factors. The treatment ameliorated memory deficits accompanied by a restoration of gamma oscillation deficits, together with a dramatic decrease in microglial activation. These findings open potential avenues for therapeutic exploration of pro-resolving LMs in AD, using a non-invasive route.


Subject(s)
Alzheimer Disease , Administration, Intranasal , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides , Animals , Inflammation , Mice
11.
Prog Neurobiol ; 210: 102213, 2022 03.
Article in English | MEDLINE | ID: mdl-34954329

ABSTRACT

Gamma oscillations (γ-oscillations) in hippocampal area CA3 are essential for memory function. Particularly, CA3 is involved in the memory related process pattern completion, which is linked with the γ-oscillations in human hippocampus. Recent studies suggest that heterogeneity in the functional properties of pyramidal cells (PCs) in CA3 plays an important role in hippocampal function. By performing concomitant recordings of PC activity and network γ-oscillations in CA3 we found three functionally-different PC subpopulations. PCs with high spike-frequency adaptation (hAPC) have the strongest action potential gamma phase-coupling, PCs with low adaptation (lAPC) show lower phase-coupling and PCs displaying a burst-firing pattern (BPC) remained quiescent. In addition, we discovered that hAPC display the highest excitatory/inhibitory drive, followed by lAPC, and lastly BPC. In conclusion, our data advance the hypothesis that PCs in CA3 are organized into subpopulations with distinct functional roles for cognition-relevant network dynamics and provide new insights in the physiology of hippocampus.


Subject(s)
CA3 Region, Hippocampal , Pyramidal Cells , Action Potentials/physiology , Animals , CA3 Region, Hippocampal/physiology , Hippocampus , Humans , Interneurons/physiology , Mice
12.
Neurobiol Dis ; 159: 105514, 2021 11.
Article in English | MEDLINE | ID: mdl-34555537

ABSTRACT

Synchronized and properly balanced electrical activity of neurons is the basis for the brain's ability to process information, to learn, and to remember. In Alzheimer's disease (AD), which causes cognitive decline in patients, this synchronization and balance is disturbed by the accumulation of neuropathological biomarkers such as amyloid-beta peptide (Aß42). Failure of Aß42 clearance mechanisms as well as desynchronization of crucial neuronal classes such as fast-spiking interneurons (FSN) are root causes for the disruption of the cognition-relevant gamma brain rhythm (30-80 Hz) and consequent cognitive impairment observed in AD. Here we show that recombinant BRICHOS molecular chaperone domains from ProSP-C or Bri2, which interfere with Aß42 aggregation, can rescue the gamma rhythm. We demonstrate that Aß42 progressively decreases gamma oscillation power and rhythmicity, disrupts the inhibition/excitation balance in pyramidal cells, and desynchronizes FSN firing during gamma oscillations in the hippocampal CA3 network of mice. Application of the more efficacious Bri2 BRICHOS chaperone rescued the cellular and neuronal network performance from all ongoing Aß42-induced functional impairments. Collectively, our findings offer critical missing data to explain the importance of FSN for normal network function and underscore the therapeutic potential of Bri2 BRICHOS to rescue the disruption of cognition-relevant brain rhythms in AD.


Subject(s)
Action Potentials/drug effects , Adaptor Proteins, Signal Transducing/pharmacology , Hippocampus/drug effects , Interneurons/drug effects , Molecular Chaperones/pharmacology , Pyramidal Cells/drug effects , Action Potentials/physiology , Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Peptides , Animals , Disease Models, Animal , Gamma Rhythm , Hippocampus/physiopathology , In Vitro Techniques , Interneurons/physiology , Mice , Neural Pathways/drug effects , Neural Pathways/physiopathology , Peptide Fragments , Protein Domains , Pulmonary Surfactant-Associated Protein C/metabolism , Pulmonary Surfactant-Associated Protein C/pharmacology , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Recombinant Proteins
13.
Mol Psychiatry ; 26(10): 5557-5567, 2021 10.
Article in English | MEDLINE | ID: mdl-34385602

ABSTRACT

In Alzheimer's disease (AD) the accumulation of amyloid-ß (Aß) correlates with degradation of cognition-relevant gamma oscillations. The gamma rhythm relies on proper neuronal spike-gamma coupling, specifically of fast-spiking interneurons (FSN). Here we tested the hypothesis that decrease in gamma power and FSN synchrony precede amyloid plaque deposition and cognitive impairment in AppNL-G-F knock-in mice (AppNL-G-F). The aim of the study was to evaluate the amyloidogenic pathology progression in the novel AppNL-G-F mouse model using in vitro electrophysiological network analysis. Using patch clamp of FSNs and pyramidal cells (PCs) with simultaneous gamma oscillation recordings, we compared the activity of the hippocampal network of wild-type mice (WT) and the AppNL-G-F mice at four disease stages (1, 2, 4, and 6 months of age). We found a severe degradation of gamma oscillation power that is independent of, and precedes Aß plaque formation, and the cognitive impairment reported previously in this animal model. The degradation correlates with increased Aß1-42 concentration in the brain. Analysis on the cellular level showed an impaired spike-gamma coupling of FSN from 2 months of age that correlates with the degradation of gamma oscillations. From 6 months of age PC firing becomes desynchronized also, correlating with reports in the literature of robust Aß plaque pathology and cognitive impairment in the AppNL-G-F mice. This study provides evidence that impaired FSN spike-gamma coupling is one of the earliest functional impairment caused by the amyloidogenic pathology progression likely is the main cause for the degradation of gamma oscillations and consequent cognitive impairment. Our data suggests that therapeutic approaches should be aimed at restoring normal FSN spike-gamma coupling and not just removal of Aß.


Subject(s)
Alzheimer Disease , Mobile Applications , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Gene Knock-In Techniques , Interneurons , Mice , Mice, Inbred C57BL , Mice, Transgenic
14.
Exp Neurol ; 343: 113743, 2021 09.
Article in English | MEDLINE | ID: mdl-34000250

ABSTRACT

Despite the development of multiple pharmacological approaches over the years aimed at treating Alzheimer's Disease (AD) only very few have been approved for clinical use in patients. To date there still exists no disease-modifying treatment that could prevent or rescue the cognitive impairment, particularly of memory aquisition, that is characteristic of AD. One of the possibilities for this state of affairs might be that the majority of drug discovery efforts focuses on outcome measures of decreased neuropathological biomarkers characteristic of AD, without taking into acount neuronal processes essential to the generation and maintenance of memory processes. Particularly, the capacity of the brain to generate theta (θ) and gamma (γ) oscillatory activity has been strongly correlated to memory performance. Using a systematic review approach, we synthesize the existing evidence in the literature on pharmacological interventions that enhance neuronal theta (θ) and/or gamma (γ) oscillations in non-pathological animal models and in AD animal models. Additionally, we synthesize the main outcomes and neurochemical systems targeted. We propose that functional biomarkers such as cognition-relevant neuronal network oscillations should be used as outcome measures during the process of research and development of novel drugs against cognitive impairment in AD.


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Gamma Rhythm/drug effects , Nerve Net/drug effects , Nootropic Agents/administration & dosage , Theta Rhythm/drug effects , Alzheimer Disease/physiopathology , Animals , Brain/physiology , Cholinergic Agents/administration & dosage , Dopamine Agents/administration & dosage , Drug Evaluation, Preclinical/methods , Electroencephalography/drug effects , Electroencephalography/methods , Gamma Rhythm/physiology , Humans , Memory Disorders/drug therapy , Memory Disorders/physiopathology , Nerve Net/physiology , Theta Rhythm/physiology , Treatment Outcome
15.
Transl Psychiatry ; 11(1): 212, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33837176

ABSTRACT

Gamma and theta brain rhythms play important roles in cognition and their interaction can affect gamma oscillation features. Hippocampal theta oscillations depend on cholinergic and GABAergic input from the medial septum-diagonal band of Broca. These projecting neurons undergo degeneration during aging and maintain high levels of neurotrophin receptor p75 (p75NTR). p75NTR mediates both apoptosis and survival and its expression is increased in Alzheimer's disease (AD) patients. Here, we investigate the importance of p75NTR for the cholinergic input to the hippocampus. Performing extracellular recordings in brain slices from p75NTR knockout mice (p75-/-) in presence of the muscarinic agonist carbachol, we find that gamma oscillation power and rhythmicity are increased compared to wild-type (WT) mice. Furthermore, gamma activity is more phase-locked to the underlying theta rhythm, which renders a stronger coupling of both rhythms. On the cellular level, we find that fast-spiking interneurons (FSNs) fire more synchronized to a preferred gamma phase in p75-/- mice. The excitatory input onto FSN is more rhythmic displaying a higher similarity with the concomitant gamma rhythm. Notably, the ablation of p75NTR counteracts the Aß-induced degradation of gamma oscillations and its nesting within the underlying theta rhythm. Our results show that the lack of p75NTR signaling could promote stronger cholinergic modulation of the hippocampal gamma rhythm, suggesting an involvement of p75NTR in the downregulation of cognition-relevant hippocampal network dynamics in pathologies. Moreover, functional data provided here suggest p75NTR as a suitable target in the search for efficacious treatments to counteract the loss of cognitive function observed in amyloid-driven pathologies such as AD.


Subject(s)
Gamma Rhythm , Theta Rhythm , Animals , Hippocampus , Humans , Mice , Mice, Knockout , Neurons
16.
J Physiol ; 598(17): 3711-3725, 2020 09.
Article in English | MEDLINE | ID: mdl-32638407

ABSTRACT

KEY POINTS: Gamma oscillations (30-80 Hz) are important for cognitive functions and depend on the synchronized activity of fast-spiking interneurons (FSN), which is crucial for network stability. Gamma oscillations are degraded in Alzheimer's disease (AD) patients exhibiting cognitive impairment, with the degree of cognitive decline correlating with the severity of gamma disruption in response to neurotoxic amyloid-beta peptide (Aß). Small molecule compounds EX15 and RE01 modulate Kv3.1/Kv3.2 potassium channels on FSN, resulting in faster activation kinetics and increased firing frequency, suggesting direct consequences for cognition-relevant gamma oscillations, particularly in a situation where network activity is pathologically compromised in the presence of neurotoxic Aß. Using electrophysiological techniques in an in vitro AD model, we found a significant effect of EX15 and RE01 with respect to counteracting toxic Aß effects on neuronal dynamics, advocating for targeting FSN activity to rescue cognitive performance from impairment caused by neurodegenerative triggers. ABSTRACT: Rhythmic electrical activity in neuronal networks such as gamma oscillations (30-80 Hz) underlies cognitive functions such as sensory perception, attention and memory. Gamma oscillations are disrupted in Alzheimer's disease (AD) patients and animal AD models, with the severity of cognitive decline correlating with the degree of rhythm disruption. Misfolded amyloid-ß peptide (Aß) is assumed to be a key trigger of AD pathology and has been show to de-synchronize action potential firing in fast-spiking interneurons (FSN), which is crucial for entraining neuronal network activity into the gamma rhythm. The synchronizing activity of FSN therefore has become one of the most suitable targets to counteract disease-driven degradation of gamma oscillations and consequent cognitive decline. EX15 and RE01 are small-molecule compounds that modulate Kv3.1/Kv3.2 potassium channels, resulting in faster activation kinetics and increased FSN firing frequency. In the present study, we investigated the potential pro-cognitive effects of EX15 and RE01 by testing their ability to modulate FSN activity during ongoing gamma oscillations in normal and Aß-disrupted network states in mouse hippocampus in vitro. In the compromised, but not the uncompromised, network state with gamma oscillations partially disrupted by Aß, both compounds improve gamma oscillation regularity by promoting re-synchronization of FSN action potential firing. Our data suggest a therapeutic potential for compounds such as EX15 and RE01, which can rescue normal action potential firing parameters in FSN, in the search for disease-modifying drug candidates counteracting the progressive dysfunction of neuronal network dynamics that underlies the cognitive impairment typical of AD and other cognitive brain disorders.


Subject(s)
Alzheimer Disease , Action Potentials , Animals , Humans , Interneurons , Mice , Mice, Inbred C57BL , Shaw Potassium Channels
17.
Neuropharmacology ; 170: 108070, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32229140

ABSTRACT

The majority of the fibroblast growth factor receptor 1-serotonin 1 A receptor (FGFR1-5-HT1AR) heterocomplexes in the hippocampus appeared to be located mainly in the neuronal networks and a relevant target for antidepressant drugs. Through a neurochemical and electrophysiological analysis it was therefore tested in the current study if astrocytic FGFR1-5-HT1AR heterocomplexes also exist in hippocampus. They may modulate the structure and function of astroglia in the hippocampus leading to possible changes in the gamma oscillations. Localization of hippocampal FGFR1-5-HT1AR heterocomplexes in astrocytes was found using in situ proximity ligation assay combined with immunohistochemistry using glial fibrillary acidic protein (GFAP) immunoreactivity as a marker for astroglia. Acute i.c.v. treatment with 8-OH-DPAT alone or together with basic fibroblast growth factor (FGF2) significantly increased FGFR1-5-HT1AR heterocomplexes in the GFAP positive cells, especially in the polymorphic layer of the dentate gyrus (PoDG) but also in the CA3 area upon combined treatment. No other hippocampal regions were studied. Also, structural plasticity changes were observed in the astrocytes, especially in the PoDG region, upon these pharmacological treatments. They may also be of relevance for enhancing the astroglial volume transmission with increased modulation of the neuronal networks in the regions studied. The effects of combined FGF2 and 5-HT agonist treatments on gamma oscillations point to a significant antagonistic interaction in astroglial FGFR1-5-HT1AR heterocomplexes that may contribute to counteraction of the 5-HT1AR-mediated decrease of gamma oscillations. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.


Subject(s)
Astrocytes/physiology , Fibroblast Growth Factor 2/physiology , Gamma Rhythm/physiology , Hippocampus/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Serotonin, 5-HT1A/physiology , Serotonin/physiology , Animals , Astrocytes/drug effects , Gamma Rhythm/drug effects , Hippocampus/drug effects , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/agonists , Serotonin 5-HT1 Receptor Agonists/pharmacology
18.
Commun Biol ; 3(1): 32, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959875

ABSTRACT

Molecular chaperones play important roles in preventing protein misfolding and its potentially harmful consequences. Deterioration of molecular chaperone systems upon ageing are thought to underlie age-related neurodegenerative diseases, and augmenting their activities could have therapeutic potential. The dementia relevant domain BRICHOS from the Bri2 protein shows qualitatively different chaperone activities depending on quaternary structure, and assembly of monomers into high-molecular weight oligomers reduces the ability to prevent neurotoxicity induced by the Alzheimer-associated amyloid-ß peptide 1-42 (Aß42). Here we design a Bri2 BRICHOS mutant (R221E) that forms stable monomers and selectively blocks a main source of toxic species during Aß42 aggregation. Wild type Bri2 BRICHOS oligomers are partly disassembled into monomers in the presence of the R221E mutant, which leads to potentiated ability to prevent Aß42 toxicity to neuronal network activity. These results suggest that the activity of endogenous molecular chaperones may be modulated to enhance anti-Aß42 neurotoxic effects.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Hippocampus/metabolism , Molecular Chaperones/metabolism , Amyloid/metabolism , Amyloid/ultrastructure , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Hippocampus/drug effects , Kinesis , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/pharmacology , Protein Aggregates/drug effects , Protein Binding , Protein Conformation , Protein Multimerization , Structure-Activity Relationship
19.
Hippocampus ; 30(2): 101-113, 2020 02.
Article in English | MEDLINE | ID: mdl-31313871

ABSTRACT

Gamma oscillations (30-80 Hz) are fast network activity patterns frequently linked to cognition. They are commonly studied in hippocampal brain slices in vitro, where they can be evoked via pharmacological activation of various receptor families. One limitation of this approach is that neuronal activity is studied in a highly artificial extracellular fluid environment, as provided by artificial cerebrospinal fluid (aCSF). Here, we examine the influence of human cerebrospinal fluid (hCSF) on kainate-evoked and spontaneous gamma oscillations in mouse hippocampus. We show that hCSF, as compared to aCSF of matched electrolyte and glucose composition, increases the power of kainate-evoked gamma oscillations and induces spontaneous gamma activity in areas CA3 and CA1 that is reversed by washout. Bath application of atropine entirely abolished hCSF-induced gamma oscillations, indicating critical contribution from muscarinic acetylcholine receptor-mediated signaling. In separate whole-cell patch clamp recordings from rat hippocampus, hCSF increased theta resonance frequency and strength in pyramidal cells along with enhancement of h-current (Ih ) amplitude. We found no evidence of intrinsic gamma frequency resonance at baseline (aCSF) among fast-spiking interneurons, and this was not altered by hCSF. However, hCSF increased the excitability of fast-spiking interneurons, which likely contributed to gamma rhythmogenesis. Our findings show that hCSF promotes network gamma oscillations in the hippocampus in vitro and suggest that neuromodulators distributed in CSF could have significant influence on neuronal network activity in vivo.


Subject(s)
Cerebrospinal Fluid , Gamma Rhythm/drug effects , Hippocampus/drug effects , Interneurons/drug effects , Pyramidal Cells/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gamma Rhythm/physiology , Hippocampus/physiology , Humans , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Interneurons/physiology , Kainic Acid/pharmacology , Mice , Patch-Clamp Techniques , Pyramidal Cells/physiology
20.
Mol Neurobiol ; 57(2): 1170-1185, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31701438

ABSTRACT

The vanilloid compound capsaicin (Cp) is best known to bind to and activate the transient receptor potential vanilloid receptor-1 (TrpV1). A growing number of studies use capsaicin as a tool to study the role of TrpV1 in the central nervous system (CNS). Although most of capsaicin's CNS effects have been reported to be mediated by TrpV1 activation, evidence exists that capsaicin can also trigger functional changes in hippocampal activity independently of TrpV1. Recently, we have reported that capsaicin induces impairment in hippocampal gamma oscillations via a TrpV1-independent pathway. Here, we dissect the underlying mechanisms of capsaicin-induced alterations to functional network dynamics. We found that capsaicin induces a reduction in action potential (AP) firing rate and a subsequent loss of synchronicity in pyramidal cell (PC) spiking activity in hippocampus. Moreover, capsaicin induces alterations in PC spike-timing since increased first-spike latency was observed after capsaicin treatment. First-spike latency can be regulated by the voltage-dependent potassium current D (ID) or Na+/K+-ATPase. Selective inhibition of ID via low 4-AP concentration and Na+/K+-ATPase using its blocker ouabain, we found that capsaicin effects on AP spike timing were completely inhibited by ouabain but not with 4-AP. In conclusion, our study shows that capsaicin in a TrpV1-independent manner and possibly involving Na+/K+-ATPase activity can impair cognition-relevant functional network dynamics such as gamma oscillations and provides important data regarding the use of capsaicin as a tool to study TrpV1 function in the CNS.


Subject(s)
Capsaicin/pharmacology , Hippocampus/drug effects , Sodium-Potassium-Exchanging ATPase/drug effects , TRPV Cation Channels/drug effects , Action Potentials/drug effects , Animals , Hippocampus/metabolism , Male , Mice , Pyramidal Cells/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , TRPV Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...