Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biol Sex Differ ; 14(1): 59, 2023 09 16.
Article in English | MEDLINE | ID: mdl-37716988

ABSTRACT

BACKGROUND: The amyloid-ß (Aß) cascade is one of the most studied theories linked to AD. In multiple models, Aß accumulation and dyshomeostasis have shown a key role in AD onset, leading to excitatory/inhibitory imbalance, the impairments of synaptic plasticity and oscillatory activity, and memory deficits. Despite the higher prevalence of Alzheimer's disease (AD) in women compared to men, the possible sex difference is scarcely explored and the information from amyloidosis transgenic mice models is contradictory. Thus, given the lack of data regarding the early stages of amyloidosis in female mice, the aim of this study was to systematically characterize the effect of an intracerebroventricular (icv.) injection of Aß1-42 on hippocampal-dependent memory, and on associated activity-dependent synaptic plasticity in the hippocampal CA1-CA3 synapse, in both male and female mice. METHODS: To do so, we evaluated long term potentiation (LTP) with ex vivo electrophysiological recordings as well as encoding and retrieval of spatial (working, short- and long-term) and exploratory habituation memories using Barnes maze and object location, or open field habituation tasks, respectively. RESULTS: Aß1-42 administration impaired all forms of memory evaluated in this work, regardless of sex. This effect was displayed in a long-lasting manner (up to 17 days post-injection). LTP was inhibited at a postsynaptic level, both in males and females, and a long-term depression (LTD) was induced for the same prolonged period, which could underlie memory deficits. CONCLUSIONS: In conclusion, our results provide further evidence on the shifting of LTP/LTD threshold due to a single icv. Aß1-42 injection, which underly cognitive deficits in the early stages of AD. These long-lasting cognitive and functional alterations in males and females validate this model for the study of early amyloidosis in both sexes, thus offering a solid alternative to the inconsistence of amyloidosis transgenic mice models.


This study focuses on investigating how amyloid-ß (Aß), a key toxic protein in Alzheimer's disease (AD), impacts memory and functioning of the synapses in both male and female mice.Our primary objective was to comprehensively understand the impact of Aß1­42, a specific form of Aß, when introduced into the brain's ventricles, focusing on memory processes associated with the hippocampus, a brain region vital for learning and memory.Prior research established Aß's significance in AD and memory decline. However, despite the higher prevalence of AD in females, the connection between Aß, memory, and sex differences required further exploration. Furthermore, findings from experiments utilizing Aß transgenic mice have offered conflicting outcomes. Here, by administering Aß1­42 to male and female mice, we systematically assessed memory using cognitive tasks. Results were consistent: memory deficits were evident in both sexes, persisting for up to 17 days post-injection.Delving deeper, we explored alterations in synaptic plasticity, a cornerstone of learning and memory. Our investigations unveiled disruptions in long-term potentiation (LTP) and long-term depression (LTD)­essential synaptic processes­in both male and female mice subjected to Aß1­42 injection.These intriguing findings underscore Aß1­42's lasting influence on memory and synaptic function, emphasizing its role in early AD-related cognitive decline. Additionally, our study highlights the potential of this experimental model to investigate early AD across sex differences, offering a promising alternative to the existing array Aß transgenic mouse models and addressing the need for a more consistent investigative framework.


Subject(s)
Alzheimer Disease , Amyloidosis , Female , Male , Humans , Mice , Animals , Neuronal Plasticity , Mice, Transgenic , Memory Disorders
2.
Neuropharmacology ; 238: 109668, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37474000

ABSTRACT

Learning and memory occurrence requires of hippocampal long-term synaptic plasticity and precise neural activity orchestrated by brain network oscillations, both processes reciprocally influencing each other. As G-protein-gated inwardly rectifying potassium (GIRK) channels rule synaptic plasticity that supports hippocampal-dependent memory, here we assessed their unknown role in hippocampal oscillatory activity in relation to synaptic plasticity induction. In alert male mice, pharmacological GIRK modulation did not alter neural oscillations before long-term potentiation (LTP) induction. However, after an LTP generating protocol, both gain- and loss-of basal GIRK activity transformed LTP into long-term depression, but only specific suppression of constitutive GIRK activity caused a disruption of network synchronization (δ, α, γ bands), even leading to long-lasting ripples and fast ripples pathological oscillations. Together, our data showed that constitutive GIRK activity plays a key role in the tuning mechanism of hippocampal oscillatory activity during long-term synaptic plasticity processes that underlies hippocampal-dependent cognitive functions.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels , Long-Term Potentiation , Mice , Male , Animals , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Hippocampus/metabolism , Neuronal Plasticity , Learning
3.
J Physiol ; 601(16): 3533-3556, 2023 08.
Article in English | MEDLINE | ID: mdl-37309891

ABSTRACT

Carnitine palmitoyltransferase 1c (CPT1C) is a neuron-specific protein widely distributed throughout the CNS and highly expressed in discrete brain areas including the hypothalamus, hippocampus, amygdala and different motor regions. Its deficiency has recently been shown to disrupt dendritic spine maturation and AMPA receptor synthesis and trafficking in the hippocampus, but its contribution to synaptic plasticity and cognitive learning and memory processes remains mostly unknown. Here, we aimed to explore the molecular, synaptic, neural network and behavioural role of CPT1C in cognition-related functions by using CPT1C knockout (KO) mice. CPT1C-deficient mice showed extensive learning and memory deficits. The CPT1C KO animals exhibited impaired motor and instrumental learning that seemed to be related, in part, to locomotor deficits and muscle weakness but not to mood alterations. In addition, CPT1C KO mice showed detrimental hippocampus-dependent spatial and habituation memory, most probably attributable to inefficient dendritic spine maturation, impairments in long-term plasticity at the CA3-CA1 synapse and aberrant cortical oscillatory activity. In conclusion, our results reveal that CPT1C is not only crucial for motor function, coordination and energy homeostasis, but also has a crucial role in the maintenance of learning and memory cognitive functions. KEY POINTS: CPT1C, a neuron-specific interactor protein involved in AMPA receptor synthesis and trafficking, was found to be highly expressed in the hippocampus, amygdala and various motor regions. CPT1C-deficient animals exhibited energy deficits and impaired locomotion, but no mood changes were found. CPT1C deficiency disrupts hippocampal dendritic spine maturation and long-term synaptic plasticity and reduces cortical γ oscillations. CPT1C was found to be crucial for motor, associative and non-associative learning and memory.


Subject(s)
Carnitine O-Palmitoyltransferase , Receptors, AMPA , Animals , Mice , Brain/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Hippocampus/metabolism , Long-Term Potentiation , Mice, Knockout , Neuronal Plasticity , Neurons/metabolism , Receptors, AMPA/genetics , Receptors, AMPA/metabolism
4.
J Neurosci ; 41(33): 7086-7102, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34261700

ABSTRACT

The G-protein-gated inwardly rectifying potassium (Kir3/GIRK) channel is the effector of many G-protein-coupled receptors (GPCRs). Its dysfunction has been linked to the pathophysiology of Down syndrome, Alzheimer's and Parkinson's diseases, psychiatric disorders, epilepsy, drug addiction, or alcoholism. In the hippocampus, GIRK channels decrease excitability of the cells and contribute to resting membrane potential and inhibitory neurotransmission. Here, to elucidate the role of GIRK channels activity in the maintenance of hippocampal-dependent cognitive functions, their involvement in controlling neuronal excitability at different levels of complexity was examined in C57BL/6 male mice. For that purpose, GIRK activity in the dorsal hippocampus CA3-CA1 synapse was pharmacologically modulated by two drugs: ML297, a GIRK channel opener, and Tertiapin-Q (TQ), a GIRK channel blocker. Ex vivo, using dorsal hippocampal slices, we studied the effect of pharmacological GIRK modulation on synaptic plasticity processes induced in CA1 by Schaffer collateral stimulation. In vivo, we performed acute intracerebroventricular (i.c.v.) injections of the two GIRK modulators to study their contribution to electrophysiological properties and synaptic plasticity of dorsal hippocampal CA3-CA1 synapse, and to learning and memory capabilities during hippocampal-dependent tasks. We found that pharmacological disruption of GIRK channel activity by i.c.v. injections, causing either function gain or function loss, induced learning and memory deficits by a mechanism involving neural excitability impairments and alterations in the induction and maintenance of long-term synaptic plasticity processes. These results support the contention that an accurate control of GIRK activity must take place in the hippocampus to sustain cognitive functions.SIGNIFICANCE STATEMENT Cognitive processes of learning and memory that rely on hippocampal synaptic plasticity processes are critically ruled by a finely tuned neural excitability. G-protein-gated inwardly rectifying K+ (GIRK) channels play a key role in maintaining resting membrane potential, cell excitability and inhibitory neurotransmission. Here, we demonstrate that modulation of GIRK channels activity, causing either function gain or function loss, transforms high-frequency stimulation (HFS)-induced long-term potentiation (LTP) into long-term depression (LTD), inducing deficits in hippocampal-dependent learning and memory. Together, our data show a crucial GIRK-activity-mediated mechanism that governs synaptic plasticity direction and modulates subsequent hippocampal-dependent cognitive functions.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Hippocampus/physiology , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Animals , Conditioning, Operant/physiology , Emotions/physiology , Excitatory Postsynaptic Potentials/physiology , Learning/physiology , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Psychomotor Performance/physiology
5.
Biology (Basel) ; 9(7)2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32698467

ABSTRACT

In early Alzheimer disease (AD) models synaptic failures and upstreaming aberrant patterns of network synchronous activity result in hippocampal-dependent memory deficits. In such initial stage, soluble forms of Amyloid-ß (Aß) peptides have been shown to play a causal role. Among different Aß species, Aß25-35 has been identified as the biologically active fragment, as induces major neuropathological signs related to early AD stages. Consequently, it has been extensively used to acutely explore the pathophysiological events related with neuronal dysfunction induced by soluble Aß forms. However, the synaptic mechanisms underlying its toxic effects on hippocampal-dependent memory remain unresolved. Here, in an in vivo model of amyloidosis generated by intracerebroventricular injections of Aß25-35 we studied the synaptic dysfunction mechanisms underlying hippocampal cognitive deficits. At the synaptic level, long-term potentiation (LTP) of synaptic excitation and inhibition was induced in CA1 region by high frequency simulation (HFS) applied to Schaffer collaterals. Aß25-35 was found to alter metaplastic mechanisms of plasticity, facilitating long-term depression (LTD) of both types of LTP. In addition, aberrant synchronization of hippocampal network activity was found while at the behavioral level, deficits in hippocampal-dependent habituation and recognition memories emerged. Together, our results provide a substrate for synaptic disruption mechanism underlying hippocampal cognitive deficits present in Aß25-35 amyloidosis model.

7.
J Neurochem ; 153(3): 362-376, 2020 05.
Article in English | MEDLINE | ID: mdl-31875959

ABSTRACT

Hippocampal synaptic plasticity disruption by amyloid-ß (Aß) peptides + thought to be responsible for learning and memory impairments in Alzheimer's disease (AD) early stage. Failures in neuronal excitability maintenance seems to be an underlying mechanism. G-protein-gated inwardly rectifying potassium (GirK) channels control neural excitability by hyperpolarization in response to many G-protein-coupled receptors activation. Here, in early in vitro and in vivo amyloidosis mouse models, we study whether GirK channels take part of the hippocampal synaptic plasticity impairments generated by Aß1-42 . In vitro electrophysiological recordings from slices showed that Aß1-42 alters synaptic plasticity by switching high-frequency stimulation (HFS) induced long-term potentiation (LTP) to long-term depression (LTD), which led to in vivo hippocampal-dependent memory deficits. Remarkably, selective pharmacological activation of GirK channels with ML297 rescued both HFS-induced LTP and habituation memory from Aß1-42 action. Moreover, when GirK channels were specifically blocked by Tertiapin-Q, their activation with ML297 failed to rescue LTP from the HFS-dependent LTD induced by Aß1-42 . On the other hand, the molecular analysis of the recorded slices by western blot showed that the expression of GIRK1/2 subunits, which form the prototypical GirK channel in the hippocampus, was not significantly regulated by Aß1-42 . However, immunohistochemical examination of our in vivo amyloidosis model showed Aß1-42 to down-regulate hippocampal GIRK1 subunit expression. Together, our results describe an Aß-mediated deleterious synaptic mechanism that modifies the induction threshold for hippocampal LTP/LTD and underlies memory alterations observed in amyloidosis models. In this scenario, GirK activation assures memory formation by preventing the transformation of HFS-induced LTP into LTD.


Subject(s)
Amyloidosis/metabolism , Excitatory Postsynaptic Potentials/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Hippocampus/metabolism , Long-Term Synaptic Depression/physiology , Memory Disorders/metabolism , Amyloid beta-Peptides/toxicity , Amyloidosis/chemically induced , Animals , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Long-Term Synaptic Depression/drug effects , Male , Memory Disorders/chemically induced , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Peptide Fragments/toxicity , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, G-Protein-Coupled/metabolism
8.
Brain Struct Funct ; 224(8): 2717-2731, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31375981

ABSTRACT

Medial rectus motoneurons are innervated by two main pontine inputs. The specific function of each of these two inputs remains to be fully understood. Indeed, selective partial deafferentation of medial rectus motoneurons, performed by the lesion of either the vestibular or the abducens input, initially induces similar changes in motoneuronal discharge. However, at longer time periods, the responses to both lesions are dissimilar. Alterations on eye movements and motoneuronal discharge induced by vestibular input transection recover completely 2 months post-lesion, whereas changes induced by abducens internuclear lesion are more drastic and permanent. Functional recovery could be due to some kind of plastic process, such as reactive synaptogenesis, developed by the remaining intact input, which would occupy the vacant synaptic spaces left after lesion. Herein, by means of confocal microscopy, immunocytochemistry and retrograde labeling, we attempt to elucidate the possible plastic processes that take place after partial deafferentation of medial rectus motoneuron. 48 h post-injury, both vestibular and abducens internuclear lesions produced a reduced synaptic coverage on these motoneurons. However, 96 h after vestibular lesion, there was a partial recovery in the number of synaptic contacts. This suggests that there was reactive synaptogenesis. This recovery was preceded by an increase in somatic neurotrophin content, suggesting a role of these molecules in presynaptic axonal sprouting. The rise in synaptic coverage might be due to terminal sprouting performed by the remaining main input, i.e., abducens internuclear neurons. The present results may improve the understanding of this apparently redundant input system.


Subject(s)
Motor Neurons/physiology , Neuronal Plasticity , Oculomotor Muscles/physiology , Animals , Denervation/methods , Eye Movements , Male , Motor Neurons/cytology , Neural Pathways/physiology , Oculomotor Muscles/innervation , Rats, Wistar
9.
Sci Rep ; 7(1): 14658, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116174

ABSTRACT

The hippocampus plays a critical role in learning and memory. Its correct performance relies on excitatory/inhibitory synaptic transmission balance. In early stages of Alzheimer's disease (AD), neuronal hyperexcitability leads to network dysfunction observed in cortical regions such as the hippocampus. G-protein-gated potassium (GirK) channels induce neurons to hyperpolarize, contribute to the resting membrane potential and could compensate any excesses of excitation. Here, we have studied the relationship between GirK channels and hippocampal function in a mouse model of early AD pathology. Intracerebroventricular injections of amyloid-ß (Aß 1-42) peptide-which have a causal role in AD pathogenesis-were performed to evaluate CA3-CA1 hippocampal synapse functionality in behaving mice. Aß increased the excitability of the CA3-CA1 synapse, impaired long-term potentiation (LTP) and hippocampal oscillatory activity, and induced deficits in novel object recognition (NOR) tests. Injection of ML297 alone, a selective GirK activator, was also translated in LTP and NOR deficits. However, increasing GirK activity rescued all hippocampal deficits induced by Aß due to the restoration of excitability values in the CA3-CA1 synapse. Our results show a synaptic mechanism, through GirK channel modulation, for the prevention of the hyperexcitability that causally contributes to synaptic, network, and cognitive deficits found in early AD pathogenesis.


Subject(s)
Alzheimer Disease/physiopathology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Hippocampus/physiopathology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/pharmacology , Animals , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/physiopathology , Disease Models, Animal , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Hippocampus/metabolism , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Phenylurea Compounds/pharmacology , Pyrazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...