Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Aging Cell ; 22(3): e13778, 2023 03.
Article in English | MEDLINE | ID: mdl-36704841

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) are critical for the maturation and plasticity of glutamatergic synapses. In the hippocampus, NMDARs mainly contain GluN2A and/or GluN2B regulatory subunits. The amyloid precursor protein (APP) has emerged as a putative regulator of NMDARs, but the impact of this interaction to their function is largely unknown. By combining patch-clamp electrophysiology and molecular approaches, we unravel a dual mechanism by which APP controls GluN2B-NMDARs, depending on the life stage. We show that APP is highly abundant specifically at the postnatal postsynapse. It interacts with GluN2B-NMDARs, controlling its synaptic content and mediated currents, both in infant mice and primary neuronal cultures. Upon aging, the APP amyloidogenic-derived C-terminal fragments, rather than APP full-length, contribute to aberrant GluN2B-NMDAR currents. Accordingly, we found that the APP processing is increased upon aging, both in mice and human brain. Interfering with stability or production of the APP intracellular domain normalized the GluN2B-NMDARs currents. While the first mechanism might be essential for synaptic maturation during development, the latter could contribute to age-related synaptic impairments.


Subject(s)
Amyloid beta-Protein Precursor , Receptors, N-Methyl-D-Aspartate , Mice , Humans , Animals , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Hippocampus/metabolism , Synapses/metabolism
2.
Front Mol Neurosci ; 15: 837448, 2022.
Article in English | MEDLINE | ID: mdl-35774868

ABSTRACT

In vitro neuronal cultures are extensively used in the field of neurosciences as they represent an accessible experimental tool for neuronal genetic manipulation, time-lapse imaging, and drug screening. Optimizing the cultivation of rodent primary neuronal cultures led to the development of defined media that support the growth and maintenance of different neuronal types. Recently, a new neuronal medium, BrainPhys (BP), was formulated envisioning the mimicry of brain physiological conditions and suitability for cultured human iPSC-derived neurons and rat primary neurons. However, its advantages in mouse primary neuronal cultures and its effects in neuronal bioenergetics are yet to be demonstrated. In this study, we validated the beneficial use of BP in mouse primary neuronal cultures based on the observation that neuronal cultures in BP media showed enhanced ATP levels, which increased throughout neuronal maturation, a finding that correlates with higher mitochondrial activity and ATP production at later maturation stages, as well as an increased glycolysis response on mitochondrial inhibition and increased mitochondrial fuel flexibility. Taken together, our data demonstrate that BP medium promotes mitochondrial activity along with neuronal maturation of in vitro cultures.

3.
NPJ Parkinsons Dis ; 8(1): 51, 2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35468899

ABSTRACT

Alpha-synuclein (aSyn) is a central player in the pathogenesis of synucleinopathies due to its accumulation in typical protein aggregates in the brain. However, it is still unclear how it contributes to neurodegeneration. Type-2 diabetes mellitus is a risk factor for Parkinson's disease (PD). Interestingly, a common molecular alteration among these disorders is the age-associated increase in protein glycation. We hypothesized that glycation-induced neuronal dysfunction is a contributing factor in synucleinopathies. Here, we dissected the impact of methylglyoxal (MGO, a glycating agent) in mice overexpressing aSyn in the brain. We found that MGO-glycation potentiates motor, cognitive, olfactory, and colonic dysfunction in aSyn transgenic (Thy1-aSyn) mice that received a single dose of MGO via intracerebroventricular injection. aSyn accumulates in the midbrain, striatum, and prefrontal cortex, and protein glycation is increased in the cerebellum and midbrain. SWATH mass spectrometry analysis, used to quantify changes in the brain proteome, revealed that MGO mainly increase glutamatergic-associated proteins in the midbrain (NMDA, AMPA, glutaminase, VGLUT and EAAT1), but not in the prefrontal cortex, where it mainly affects the electron transport chain. The glycated proteins in the midbrain of MGO-injected Thy1-aSyn mice strongly correlate with PD and dopaminergic pathways. Overall, we demonstrated that MGO-induced glycation accelerates PD-like sensorimotor and cognitive alterations and suggest that the increase of glutamatergic signaling may underly these events. Our study sheds new light into the enhanced vulnerability of the midbrain in PD-related synaptic dysfunction and suggests that glycation suppressors and anti-glutamatergic drugs may hold promise as disease-modifying therapies for synucleinopathies.

4.
Mol Psychiatry ; 25(8): 1876-1900, 2020 08.
Article in English | MEDLINE | ID: mdl-29950682

ABSTRACT

Synaptic dysfunction plays a central role in Alzheimer's disease (AD), since it drives the cognitive decline. An association between a polymorphism of the adenosine A2A receptor (A2AR) encoding gene-ADORA2A, and hippocampal volume in AD patients was recently described. In this study, we explore the synaptic function of A2AR in age-related conditions. We report, for the first time, a significant overexpression of A2AR in hippocampal neurons of aged humans, which is aggravated in AD patients. A similar profile of A2AR overexpression in rats was sufficient to drive age-like memory impairments in young animals and to uncover a hippocampal LTD-to-LTP shift. This was accompanied by increased NMDA receptor gating, dependent on mGluR5 and linked to enhanced Ca2+ influx. We confirmed the same plasticity shift in memory-impaired aged rats and APP/PS1 mice modeling AD, which was rescued upon A2AR blockade. This A2AR/mGluR5/NMDAR interaction might prove a suitable alternative for regulating aberrant mGluR5/NMDAR signaling in AD without disrupting their constitutive activity.


Subject(s)
Aging/metabolism , Long-Term Synaptic Depression , Neurons/metabolism , Receptor, Adenosine A2A/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Adenosine/metabolism , Alzheimer Disease/metabolism , Animals , Cells, Cultured , Hippocampus/metabolism , Humans , Mice , Rats , Rats, Sprague-Dawley , Spatial Memory
5.
Sci Immunol ; 4(40)2019 10 11.
Article in English | MEDLINE | ID: mdl-31604844

ABSTRACT

The notion of "immune privilege" of the brain has been revised to accommodate its infiltration, at steady state, by immune cells that participate in normal neurophysiology. However, the immune mechanisms that regulate learning and memory remain poorly understood. Here, we show that noninflammatory interleukin-17 (IL-17) derived from a previously unknown fetal-derived meningeal-resident γδ T cell subset promotes cognition. When tested in classical spatial learning paradigms, mice lacking γδ T cells or IL-17 displayed deficient short-term memory while retaining long-term memory. The plasticity of glutamatergic synapses was reduced in the absence of IL-17, resulting in impaired long-term potentiation in the hippocampus. Conversely, IL-17 enhanced glial cell production of brain-derived neurotropic factor, whose exogenous provision rescued the synaptic and behavioral phenotypes of IL-17-deficient animals. Together, our work provides previously unknown clues on the mechanisms that regulate short-term versus long-term memory and on the evolutionary and functional link between the immune and nervous systems.


Subject(s)
Interleukin-17/immunology , Memory, Short-Term , Meninges/immunology , Neuronal Plasticity/immunology , T-Lymphocytes/immunology , Animals , Interleukin-17/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Brain ; 142(11): 3636-3654, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31599329

ABSTRACT

Accumulating data support the role of tau pathology in cognitive decline in ageing and Alzheimer's disease, but underlying mechanisms remain ill-defined. Interestingly, ageing and Alzheimer's disease have been associated with an abnormal upregulation of adenosine A2A receptor (A2AR), a fine tuner of synaptic plasticity. However, the link between A2AR signalling and tau pathology has remained largely unexplored. In the present study, we report for the first time a significant upregulation of A2AR in patients suffering from frontotemporal lobar degeneration with the MAPT P301L mutation. To model these alterations, we induced neuronal A2AR upregulation in a tauopathy mouse model (THY-Tau22) using a new conditional strain allowing forebrain overexpression of the receptor. We found that neuronal A2AR upregulation increases tau hyperphosphorylation, potentiating the onset of tau-induced memory deficits. This detrimental effect was linked to a singular microglial signature as revealed by RNA sequencing analysis. In particular, we found that A2AR overexpression in THY-Tau22 mice led to the hippocampal upregulation of C1q complement protein-also observed in patients with frontotemporal lobar degeneration-and correlated with the loss of glutamatergic synapses, likely underlying the observed memory deficits. These data reveal a key impact of overactive neuronal A2AR in the onset of synaptic loss in tauopathies, paving the way for new therapeutic approaches.


Subject(s)
Complement C1q/metabolism , Neurons/metabolism , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Synapses/pathology , Tauopathies/genetics , Tauopathies/pathology , Animals , Autopsy , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Memory Disorders/etiology , Memory Disorders/psychology , Mice , Mice, Transgenic , Mutation , Spatial Learning , Tauopathies/psychology , tau Proteins/genetics
7.
Cell Rep ; 29(2): 317-331.e5, 2019 Oct 08.
Article in English | MEDLINE | ID: mdl-31597094

ABSTRACT

There is a growing consensus that Alzheimer's disease (AD) involves failure of the homeostatic machinery, which underlies the firing stability of neural circuits. What are the culprits leading to neuron firing instability? The amyloid precursor protein (APP) is central to AD pathogenesis, and we recently showed that its intracellular domain (AICD) could modify synaptic signal integration. We now hypothesize that AICD modifies neuron firing activity, thus contributing to the disruption of memory processes. Using cellular, electrophysiological, and behavioral techniques, we show that pathological AICD levels weaken CA1 neuron firing activity through a gene-transcription-dependent mechanism. Furthermore, increased AICD production in hippocampal neurons modifies oscillatory activity, specifically in the γ-frequency range, and disrupts spatial memory task. Collectively, our data suggest that AICD pathological levels, observed in AD mouse models and in human patients, might contribute to progressive neuron homeostatic failure, driving the shift from normal aging to AD.


Subject(s)
Action Potentials/physiology , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , CA1 Region, Hippocampal/physiology , Neurons/physiology , Spatial Memory/physiology , Animals , Calcium Channels/metabolism , Gamma Rhythm/physiology , Humans , Male , Mice, Inbred C57BL , Models, Biological , Potassium Channels/metabolism , Protein Domains , Rats, Sprague-Dawley , Structure-Activity Relationship , Transcription, Genetic
8.
J Caffeine Adenosine Res ; 9(3): 104-127, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31559391

ABSTRACT

While neuronal loss has long been considered as the main contributor to age-related cognitive decline, these alterations are currently attributed to gradual synaptic dysfunction driven by calcium dyshomeostasis and alterations in ionotropic/metabotropic receptors. Given the key role of the hippocampus in encoding, storage, and retrieval of memory, the morpho- and electrophysiological alterations that occur in the major synapse of this network-the glutamatergic-deserve special attention. We guide you through the hippocampal anatomy, circuitry, and function in physiological context and focus on alterations in neuronal morphology, calcium dynamics, and plasticity induced by aging and Alzheimer's disease (AD). We provide state-of-the art knowledge on glutamatergic transmission and discuss implications of these novel players for intervention. A link between regular consumption of caffeine-an adenosine receptor blocker-to decreased risk of AD in humans is well established, while the mechanisms responsible have only now been uncovered. We review compelling evidence from humans and animal models that implicate adenosine A2A receptors (A2AR) upsurge as a crucial mediator of age-related synaptic dysfunction. The relevance of this mechanism in patients was very recently demonstrated in the form of a significant association of the A2AR-encoding gene with hippocampal volume (synaptic loss) in mild cognitive impairment and AD. Novel pathways implicate A2AR in the control of mGluR5-dependent NMDAR activation and subsequent Ca2+ dysfunction upon aging. The nature of this receptor makes it particularly suited for long-term therapies, as an alternative for regulating aberrant mGluR5/NMDAR signaling in aging and disease, without disrupting their crucial constitutive activity.

9.
Nat Neurosci ; 20(11): 1569-1579, 2017 11.
Article in English | MEDLINE | ID: mdl-28945221

ABSTRACT

Synucleinopathies, such as Parkinson's disease and dementia with Lewy bodies, are neurodegenerative disorders that are characterized by the accumulation of α-synuclein (aSyn) in intracellular inclusions known as Lewy bodies. Prefibrillar soluble aSyn oligomers, rather than larger inclusions, are currently considered to be crucial species underlying synaptic dysfunction. We identified the cellular prion protein (PrPC) as a key mediator in aSyn-induced synaptic impairment. The aSyn-associated impairment of long-term potentiation was blocked in Prnp null mice and rescued following PrPC blockade. We found that extracellular aSyn oligomers formed a complex with PrPC that induced the phosphorylation of Fyn kinase via metabotropic glutamate receptors 5 (mGluR5). aSyn engagement of PrPC and Fyn activated NMDA receptor (NMDAR) and altered calcium homeostasis. Blockade of mGluR5-evoked phosphorylation of NMDAR in aSyn transgenic mice rescued synaptic and cognitive deficits, supporting the hypothesis that a receptor-mediated mechanism, independent of pore formation and membrane leakage, is sufficient to trigger early synaptic damage induced by extracellular aSyn.


Subject(s)
Cognitive Dysfunction/metabolism , PrPC Proteins/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , alpha-Synuclein/metabolism , Animals , Cells, Cultured , Cognitive Dysfunction/pathology , Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Protein Binding/physiology , Rats , Rats, Sprague-Dawley
10.
Sci Rep ; 6: 31493, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27510168

ABSTRACT

Caffeine is associated with procognitive effects in humans by counteracting overactivation of the adenosine A2A receptor (A2AR), which is upregulated in the human forebrain of aged and Alzheimer's disease (AD) patients. We have previously shown that an anti-A2AR therapy reverts age-like memory deficits, by reestablishment of the hypothalamic-pituitary-adrenal (HPA) axis feedback and corticosterone circadian levels. These observations suggest that A2AR over-activation and glucocorticoid dysfunction are key events in age-related hippocampal deficits; but their direct connection has never been explored. We now show that inducing A2AR overexpression in an aging-like profile is sufficient to trigger HPA-axis dysfunction, namely loss of plasmatic corticosterone circadian oscillation, and promotes reduction of GR hippocampal levels. The synaptic plasticity and memory deficits triggered by GR in the hippocampus are amplified by A2AR over-activation and were rescued by anti-A2AR therapy; finally, we demonstrate that A2AR act on GR nuclear translocation and GR-dependent transcriptional regulation. We provide the first demonstration that A2AR is a major regulator of GR function and that this functional interconnection may be a trigger to age-related memory deficits. This supports the idea that the procognitive effects of A2AR antagonists, namely caffeine, on Alzheimer's and age-related cognitive impairments may rely on its ability to modulate GR actions.


Subject(s)
Aging/physiology , Cognitive Dysfunction/genetics , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Receptors, Glucocorticoid/metabolism , Aging/metabolism , Animals , Caffeine/metabolism , Cell Nucleus/metabolism , Cognitive Dysfunction/metabolism , Corticosterone/metabolism , Humans , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Male , Mice, Transgenic , Models, Animal , Neuronal Plasticity , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...