Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Biophys J ; 121(4): 644-657, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-34999132

RESUMO

In this work, we highlight an electrophysiological feature often observed in recordings from mouse CA1 pyramidal cells that has so far been ignored by experimentalists and modelers. It consists of a large and dynamic increase in the depolarization baseline (i.e., the minimum value of the membrane potential between successive action potentials during a sustained input) in response to strong somatic current injections. Such an increase can directly affect neurotransmitter release properties and, more generally, the efficacy of synaptic transmission. However, it cannot be explained by any currently available conductance-based computational model. Here we present a model addressing this issue, demonstrating that experimental recordings can be reproduced by assuming that an input current modifies, in a time-dependent manner, the electrical and permeability properties of the neuron membrane by shifting the ionic reversal potentials and channel kinetics. For this reason, we propose that any detailed model of ion channel kinetics for neurons exhibiting this characteristic should be adapted to correctly represent the response and the synaptic integration process during strong and sustained inputs.


Assuntos
Hipocampo , Células Piramidais , Potenciais de Ação/fisiologia , Animais , Hipocampo/fisiologia , Camundongos , Neurônios , Transmissão Sináptica
2.
Mol Psychiatry ; 25(8): 1876-1900, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-29950682

RESUMO

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.


Assuntos
Envelhecimento/metabolismo , Depressão Sináptica de Longo Prazo , Neurônios/metabolismo , Receptor A2A de Adenosina/metabolismo , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Adenosina/metabolismo , Doença de Alzheimer/metabolismo , Animais , Células Cultivadas , Hipocampo/metabolismo , Humanos , Camundongos , Ratos , Ratos Sprague-Dawley , Memória Espacial
3.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33255764

RESUMO

Therapeutic approaches providing effective medication for Alzheimer's disease (AD) patients after disease onset are urgently needed. Previous studies in AD mouse models suggested that physical exercise or changed lifestyle can delay AD-related synaptic and memory dysfunctions when treatment started in juvenile animals long before onset of disease symptoms, while a pharmacological treatment that can reverse synaptic and memory deficits in AD mice was thus far not identified. Repurposing food and drug administration (FDA)-approved drugs for treatment of AD is a promising way to reduce the time to bring such medication into clinical practice. The sphingosine-1 phosphate analog fingolimod (FTY720) was approved recently for treatment of multiple sclerosis patients. Here, we addressed whether fingolimod rescues AD-related synaptic deficits and memory dysfunction in an amyloid precursor protein/presenilin-1 (APP/PS1) AD mouse model when medication starts after onset of symptoms (at five months). Male mice received intraperitoneal injections of fingolimod for one to two months starting at five to six months. This treatment rescued spine density as well as long-term potentiation in hippocampal cornu ammonis-1 (CA1) pyramidal neurons, that were both impaired in untreated APP/PS1 animals at six to seven months of age. Immunohistochemical analysis with markers of microgliosis (ionized calcium-binding adapter molecule 1; Iba1) and astrogliosis (glial fibrillary acid protein; GFAP) revealed that our fingolimod treatment regime strongly down regulated neuroinflammation in the hippocampus and neocortex of this AD model. These effects were accompanied by a moderate reduction of Aß accumulation in hippocampus and neocortex. Our results suggest that fingolimod, when applied after onset of disease symptoms in an APP/PS1 mouse model, rescues synaptic pathology that is believed to underlie memory deficits in AD mice, and that this beneficial effect is mediated via anti-neuroinflammatory actions of the drug on microglia and astrocytes.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Precursor de Proteína beta-Amiloide/genética , Inflamação/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico , Presenilina-1/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Anti-Inflamatórios/farmacologia , Astrócitos/metabolismo , Astrócitos/patologia , Modelos Animais de Doenças , Cloridrato de Fingolimode/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Sinapses/genética , Sinapses/patologia
4.
Neurobiol Dis ; 125: 31-44, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30659983

RESUMO

SCN1A (NaV1.1 sodium channel) mutations cause Dravet syndrome (DS) and GEFS+ (which is in general milder), and are risk factors in other epilepsies. Phenotypic variability limits precision medicine in epilepsy, and it is important to identify factors that set phenotype severity and their mechanisms. It is not yet clear whether SCN1A mutations are necessary for the development of severe phenotypes or just for promoting seizures. A relevant example is the pleiotropic R1648H mutation that can cause either mild GEFS+ or severe DS. We used a R1648H knock-in mouse model (Scn1aRH/+) with mild/asymptomatic phenotype to dissociate the effects of seizures and of the mutation per se. The induction of short repeated seizures, at the age of disease onset for Scn1a mouse models (P21), had no effect in WT mice, but transformed the mild/asymptomatic phenotype of Scn1aRH/+ mice into a severe DS-like phenotype, including frequent spontaneous seizures and cognitive/behavioral deficits. In these mice, we found no major modifications in cytoarchitecture or neuronal death, but increased excitability of hippocampal granule cells, consistent with a pathological remodeling. Therefore, we demonstrate for our model that an SCN1A mutation is a prerequisite for a long term deleterious effect of seizures on the brain, indicating a clear interaction between seizures and the mutation for the development of a severe phenotype generated by pathological remodeling. Applied to humans, this result suggests that genetic alterations, even if mild per se, may increase the risk of second hits to develop severe phenotypes.


Assuntos
Epilepsia/genética , Epilepsia/patologia , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Convulsões/genética , Convulsões/patologia , Animais , Técnicas de Introdução de Genes , Hipocampo/patologia , Camundongos , Mutação , Fenótipo
5.
Neuron ; 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38878768

RESUMO

NMDA receptors (NMDARs) are ionotropic receptors crucial for brain information processing. Yet, evidence also supports an ion-flux-independent signaling mode mediating synaptic long-term depression (LTD) and spine shrinkage. Here, we identify AETA (Aη), an amyloid-ß precursor protein (APP) cleavage product, as an NMDAR modulator with the unique dual regulatory capacity to impact both signaling modes. AETA inhibits ionotropic NMDAR activity by competing with the co-agonist and induces an intracellular conformational modification of GluN1 subunits. This favors non-ionotropic NMDAR signaling leading to enhanced LTD and favors spine shrinkage. Endogenously, AETA production is increased by in vivo chemogenetically induced neuronal activity. Genetic deletion of AETA production alters NMDAR transmission and prevents LTD, phenotypes rescued by acute exogenous AETA application. This genetic deletion also impairs contextual fear memory. Our findings demonstrate AETA-dependent NMDAR activation (ADNA), characterizing AETA as a unique type of endogenous NMDAR modulator that exerts bidirectional control over NMDAR signaling and associated information processing.

6.
Curr Opin Neurobiol ; 82: 102754, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37542943

RESUMO

The amyloid-ß precursor protein (APP) is a ubiquitous protein with a strong genetic link to Alzheimer's disease. Although the protein was identified more than forty years ago, its physiological function is still unclear. In recent years, advances in technology have allowed researchers to tackle APP functions in greater depth. In this review, we discuss the latest research pertaining to APP functions from development to aging. We also address the different roles that APP could play in specific types of cells of the central and peripheral nervous system and in other organs of the body. We argue that, until we fully identify the functions of APP in space and time, we will be missing important pieces of the puzzle to solve its pathological implication in Alzheimer's disease and beyond.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Humanos , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Sinapses/metabolismo , Sistema Nervoso Periférico/patologia , Peptídeos beta-Amiloides/metabolismo
7.
Aging Cell ; 22(3): e13778, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36704841

RESUMO

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.


Assuntos
Precursor de Proteína beta-Amiloide , Receptores de N-Metil-D-Aspartato , Camundongos , Humanos , Animais , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Hipocampo/metabolismo , Sinapses/metabolismo
8.
Front Synaptic Neurosci ; 13: 799274, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34970134

RESUMO

Drug addiction is defined as a compulsive pattern of drug-seeking- and taking- behavior, with recurrent episodes of abstinence and relapse, and a loss of control despite negative consequences. Addictive drugs promote reinforcement by increasing dopamine in the mesocorticolimbic system, which alters excitatory glutamate transmission within the reward circuitry, thereby hijacking reward processing. Within the reward circuitry, the striatum is a key target structure of drugs of abuse since it is at the crossroad of converging glutamate inputs from limbic, thalamic and cortical regions, encoding components of drug-associated stimuli and environment, and dopamine that mediates reward prediction error and incentive values. These signals are integrated by medium-sized spiny neurons (MSN), which receive glutamate and dopamine axons converging onto their dendritic spines. MSN primarily form two mostly distinct populations based on the expression of either DA-D1 (D1R) or DA-D2 (D2R) receptors. While a classical view is that the two MSN populations act in parallel, playing antagonistic functional roles, the picture seems much more complex. Herein, we review recent studies, based on the use of cell-type-specific manipulations, demonstrating that dopamine differentially modulates dendritic spine density and synapse formation, as well as glutamate transmission, at specific inputs projecting onto D1R-MSN and D2R-MSN to shape persistent pathological behavioral in response to drugs of abuse. We also discuss the identification of distinct molecular events underlying the detrimental interplay between dopamine and glutamate signaling in D1R-MSN and D2R-MSN and highlight the relevance of such cell-type-specific molecular studies for the development of innovative strategies with potential therapeutic value for addiction. Because drug addiction is highly prevalent in patients with other psychiatric disorders when compared to the general population, we last discuss the hypothesis that shared cellular and molecular adaptations within common circuits could explain the co-occurrence of addiction and depression. We will therefore conclude this review by examining how the nucleus accumbens (NAc) could constitute a key interface between addiction and depression.

9.
Alzheimers Res Ther ; 13(1): 125, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34238366

RESUMO

BACKGROUND: Amyloid precursor protein (APP) processing is central to Alzheimer's disease (AD) etiology. As early cognitive alterations in AD are strongly correlated to abnormal information processing due to increasing synaptic impairment, it is crucial to characterize how peptides generated through APP cleavage modulate synapse function. We previously described a novel APP processing pathway producing η-secretase-derived peptides (Aη) and revealed that Aη-α, the longest form of Aη produced by η-secretase and α-secretase cleavage, impaired hippocampal long-term potentiation (LTP) ex vivo and neuronal activity in vivo. METHODS: With the intention of going beyond this initial observation, we performed a comprehensive analysis to further characterize the effects of both Aη-α and the shorter Aη-ß peptide on hippocampus function using ex vivo field electrophysiology, in vivo multiphoton calcium imaging, and in vivo electrophysiology. RESULTS: We demonstrate that both synthetic peptides acutely impair LTP at low nanomolar concentrations ex vivo and reveal the N-terminus to be a primary site of activity. We further show that Aη-ß, like Aη-α, inhibits neuronal activity in vivo and provide confirmation of LTP impairment by Aη-α in vivo. CONCLUSIONS: These results provide novel insights into the functional role of the recently discovered η-secretase-derived products and suggest that Aη peptides represent important, pathophysiologically relevant, modulators of hippocampal network activity, with profound implications for APP-targeting therapeutic strategies in AD.


Assuntos
Doença de Alzheimer , Potenciação de Longa Duração , Secretases da Proteína Precursora do Amiloide , Peptídeos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Humanos , Neurônios
10.
Nat Commun ; 12(1): 1557, 2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33692361

RESUMO

Fragile X syndrome (FXS) is the most frequent form of inherited intellectual disability and the best-described monogenic cause of autism. CGG-repeat expansion in the FMR1 gene leads to FMR1 silencing, loss-of-expression of the Fragile X Mental Retardation Protein (FMRP), and is a common cause of FXS. Missense mutations in the FMR1 gene were also identified in FXS patients, including the recurrent FMRP-R138Q mutation. To investigate the mechanisms underlying FXS caused by this mutation, we generated a knock-in mouse model (Fmr1R138Q) expressing the FMRP-R138Q protein. We demonstrate that, in the hippocampus of the Fmr1R138Q mice, neurons show an increased spine density associated with synaptic ultrastructural defects and increased AMPA receptor-surface expression. Combining biochemical assays, high-resolution imaging, electrophysiological recordings, and behavioural testing, we also show that the R138Q mutation results in impaired hippocampal long-term potentiation and socio-cognitive deficits in mice. These findings reveal the functional impact of the FMRP-R138Q mutation in a mouse model of FXS.


Assuntos
Disfunção Cognitiva/genética , Disfunção Cognitiva/fisiopatologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Mutação de Sentido Incorreto/fisiologia , Receptores de Glutamato/metabolismo , Animais , Biotinilação , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Células Cultivadas , Disfunção Cognitiva/metabolismo , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Immunoblotting , Potenciação de Longa Duração/genética , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Mutação de Sentido Incorreto/genética , Técnicas de Patch-Clamp , Receptores de Glutamato/genética
11.
Sci Adv ; 7(43): eabg5970, 2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34669474

RESUMO

Addictive drugs increase dopamine in the nucleus accumbens (NAc), where it persistently shapes excitatory glutamate transmission and hijacks natural reward processing. Here, we provide evidence, from mice to humans, that an underlying mechanism relies on drug-evoked heteromerization of glutamate N-methyl-d-aspartate receptors (NMDAR) with dopamine receptor 1 (D1R) or 2 (D2R). Using temporally controlled inhibition of D1R-NMDAR heteromerization, we unraveled their selective implication in early phases of cocaine-mediated synaptic, morphological, and behavioral responses. In contrast, preventing D2R-NMDAR heteromerization blocked the persistence of these adaptations. Interfering with these heteromers spared natural reward processing. Notably, we established that D2R-NMDAR complexes exist in human samples and showed that, despite a decreased D2R protein expression in the NAc, individuals with psychostimulant use disorder display a higher proportion of D2R forming heteromers with NMDAR. These findings contribute to a better understanding of molecular mechanisms underlying addiction and uncover D2R-NMDAR heteromers as targets with potential therapeutic value.

12.
Cell Rep ; 36(9): 109574, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34469732

RESUMO

Neuroinflammation in patients with Alzheimer's disease (AD) and related mouse models has been recognized for decades, but the contribution of the recently described meningeal immune population to AD pathogenesis remains to be addressed. Here, using the 3xTg-AD model, we report an accumulation of interleukin-17 (IL-17)-producing cells, mostly γδ T cells, in the brain and the meninges of female, but not male, mice, concomitant with the onset of cognitive decline. Critically, IL-17 neutralization into the ventricles is sufficient to prevent short-term memory and synaptic plasticity deficits at early stages of disease. These effects precede blood-brain barrier disruption and amyloid-beta or tau pathology, implying an early involvement of IL-17 in AD pathology. When IL-17 is neutralized at later stages of disease, the onset of short-memory deficits and amyloidosis-related splenomegaly is delayed. Altogether, our data support the idea that cognition relies on a finely regulated balance of "inflammatory" cytokines derived from the meningeal immune system.


Assuntos
Doença de Alzheimer/metabolismo , Comportamento Animal , Encéfalo/metabolismo , Cognição , Mediadores da Inflamação/metabolismo , Interleucina-17/metabolismo , Linfócitos Intraepiteliais/metabolismo , Doenças Neuroinflamatórias/metabolismo , Sinapses/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/psicologia , Animais , Anti-Inflamatórios/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Mediadores da Inflamação/antagonistas & inibidores , Interleucina-17/antagonistas & inibidores , Linfócitos Intraepiteliais/efeitos dos fármacos , Masculino , Memória de Curto Prazo , Camundongos da Linhagem 129 , Camundongos Transgênicos , Doenças Neuroinflamatórias/patologia , Doenças Neuroinflamatórias/prevenção & controle , Doenças Neuroinflamatórias/psicologia , Plasticidade Neuronal , Sinapses/efeitos dos fármacos , Sinapses/patologia
13.
J Pharmacol Exp Ther ; 332(1): 153-63, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19789361

RESUMO

Adenosine-induced modulation of neuromuscular transmission in young (3-4-week-old) rats was evaluated. Inhibition of adenosine kinase with iodotubercidin (ITU; 10 microM), which is known to induce adenosine release, enhanced the amplitude of evoked end-plate potentials (EPPs) recorded from innervated diaphragm muscle fibers. This facilitatory effect was transformed into an inhibitory one upon blockade of adenosine A(2A) receptors with 4-(2-[7-amino-2-(2-furly)[1,2,4]triazolo[2,3-a][1,3,5]triazin5ylamino] ethyl) phenol (ZM 241385) (50 nM); further blockade of adenosine A(1) receptors with the selective antagonist 1,3-dipropyl-8-cyclopentylxanthine (DPCPX; 10 nM) abolished that inhibition. Adenosine or 2-chloroadenosine (CADO), at submicromolar concentrations, increased the amplitude and the quantal content of EPPs, whereas at low micromolar concentrations they decreased EPP amplitude. Blockade of A(1) receptors with DPCPX (10 nM) prevented both excitatory and inhibitory effects, whereas blockade of A(2A) receptors with ZM241385 (50 nM) prevented only the excitatory effects. DPCPX and ZM241385 also prevented the excitatory effect of the selective A(2A) receptor agonist 2-[p-(2-carboxyethyl) phenethylamino]-5'-N-ethylcarboxamido adenosine hydrochloride (CGS 21680; 10 nM). CADO (30 nM) also increased neuromuscular transmission in adult (12-16-week-old) rats. It is suggested that at the motor nerve endings, low extracellular concentrations of adenosine activate both A(2A) and A(1) receptors, but activation of A(2A) receptors predominates over A(1) receptors; the activity of A(2A) receptors might, however, require coactivation of A(1) receptors. This facilitatory action of low concentrations of extracellular adenosine upon acetylcholine release may be particularly relevant at developing neuromuscular junctions, where subtle changes in synaptic levels of acetylcholine might influence synaptic stabilization.


Assuntos
Adenosina/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Junção Neuromuscular/efeitos dos fármacos , 2-Cloroadenosina/farmacologia , Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Animais , Diafragma/efeitos dos fármacos , Diafragma/inervação , Diafragma/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/fisiologia , Ligantes , Masculino , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Junção Neuromuscular/fisiologia , Ratos , Ratos Wistar , Receptor A1 de Adenosina/fisiologia , Receptor A2A de Adenosina/fisiologia , Tubercidina/análogos & derivados , Tubercidina/farmacologia
14.
J Caffeine Adenosine Res ; 9(3): 104-127, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31559391

RESUMO

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.

15.
Sci Immunol ; 4(40)2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31604844

RESUMO

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.


Assuntos
Interleucina-17/imunologia , Memória de Curto Prazo , Meninges/imunologia , Plasticidade Neuronal/imunologia , Linfócitos T/imunologia , Animais , Interleucina-17/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
16.
Cell Rep ; 29(2): 317-331.e5, 2019 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-31597094

RESUMO

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.


Assuntos
Potenciais de Ação/fisiologia , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Região CA1 Hipocampal/fisiologia , Neurônios/fisiologia , Memória Espacial/fisiologia , Animais , Canais de Cálcio/metabolismo , Ritmo Gama/fisiologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Canais de Potássio/metabolismo , Domínios Proteicos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Transcrição Gênica
17.
Elife ; 62017 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-28682239

RESUMO

The amyloid precursor protein (APP) harbors physiological roles at synapses and is central to Alzheimer's disease (AD) pathogenesis. Evidence suggests that APP intracellular domain (AICD) could regulate synapse function, but the underlying molecular mechanisms remain unknown. We addressed AICD actions at synapses, per se, combining in vivo AICD expression, ex vivo AICD delivery or APP knock-down by in utero electroporation of shRNAs with whole-cell electrophysiology. We report a critical physiological role of AICD in controlling GluN2B-containing NMDA receptors (NMDARs) at immature excitatory synapses, via a transcription-dependent mechanism. We further show that AICD increase in mature neurons, as reported in AD, alters synaptic NMDAR composition to an immature-like GluN2B-rich profile. This disrupts synaptic signal integration, via over-activation of SK channels, and synapse plasticity, phenotypes rescued by GluN2B antagonism. We provide a new physiological role for AICD, which becomes pathological upon AICD increase in mature neurons. Thus, AICD could contribute to AD synaptic failure.


Assuntos
Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/patologia , Neurogênese/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Camundongos , Domínios Proteicos , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
18.
Neurosci Lett ; 404(1-2): 143-7, 2006 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-16790314

RESUMO

Motor nerve terminals possess adenosine A(2A) receptors and brain derived neurotrophic factor (BDNF) TrkB receptors. In the present work we evaluated how BDNF actions on neuromuscular transmission would be influenced by adenosine A(2A) receptors activation. BDNF (20-100 ng/ml) on its own was devoid of effect on evoked endplate potentials (EPPs) recorded intracellularly from rat innervated diaphragms paralysed with tubocurarine. However, when BDNF was applied 45 min after a brief (2 min) depolarizing KCl (10 mM) pulse or when the adenosine A(2A) receptors were activated with CGS 21680 (10 nM), BDNF (20 ng/ml) increased EPPs amplitude without influencing the resting membrane potential of the muscle fibre. The action of BDNF was prevented by the adenosine A(2A) receptor antagonist, ZM 241385 (50 nM) as well as by the TrkB receptor phosphorylation inhibitor, K252a (200 nM). The PKA inhibitor, H-89 (1 microM), prevented the excitatory effect of CGS 21680 (10 nM) on EPPs as well as prevented its ability to trigger a BDNF effect. The PLCgamma inhibitor, U73122 (5 microM), did not prevent the excitatory action of CGS 21680 (10 nM) on neuromuscular transmission, but abolished the action of BDNF in the presence of the A(2A) receptor agonist. The results suggest the following sequence of events in what concerns cooperativity between A(2A) receptors and TrkB receptors at the neuromuscular junction: A(2A) receptor activates the PKA pathway, which promotes the action of BDNF through TrkB receptors coupled to PLCgamma, leading to enhancement of neuromuscular transmission.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Junção Neuromuscular/fisiologia , Receptor A2A de Adenosina/fisiologia , Transmissão Sináptica/fisiologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina , Animais , Diafragma/efeitos dos fármacos , Diafragma/fisiologia , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Junção Neuromuscular/efeitos dos fármacos , Fenetilaminas/farmacologia , Ratos , Transmissão Sináptica/efeitos dos fármacos
19.
Neurosci Lett ; 584: 224-9, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25449868

RESUMO

At the neuromuscular junction, spontaneous giant events (GMEPPs) are enhanced in different conditions when degenerative and/or remodeling processes take place, but no one investigated their incidence upon aging. In the present work, we evaluated evoked and spontaneous neuromuscular transmission events recorded from single muscle fibers. Phrenic-diaphragm preparations of 3-4, 12-16, 36-40 and 70-80 weeks old rat males were used. We found that the occurrence of GMEPPs significantly increases in aged rats. Moreover, in old rats the neuromuscular transmission was significantly impaired due to a significant decrease in the amplitude and quantal content of evoked endplate potentials. Interestingly, the number of observed EPPs failures were ∼ 3 times lower than the predicted value based on the quantal content. This discrepancy was not observed in infant or adult rats. The coincidence of a high GMEPPs frequency with a lower than expected EPPs failure rate suggests that GMEPPs events are needed to preserve effective neuromuscular transmission in aged animals.


Assuntos
Envelhecimento/fisiologia , Potenciais Pós-Sinápticos em Miniatura , Acetilcolina/metabolismo , Animais , Diafragma/inervação , Diafragma/fisiologia , Masculino , Fibras Musculares Esqueléticas/fisiologia , Junção Neuromuscular/fisiologia , Nervo Frênico/fisiologia , Ratos Wistar
20.
PLoS One ; 9(8): e104081, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25093813

RESUMO

Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease leading to motor neuron dysfunction resulting in impairment of neuromuscular transmission. A2A adenosine receptors have already been considered as a potential therapeutical target for ALS but their neuromodulatory role at the neuromuscular junction in ALS remains to be clarified. In the present work, we evaluated the effects of A2A receptors on neuromuscular transmission of an animal model of ALS: SOD1(G93A) mice either in the pre-symptomatic (4-6 weeks old) or in the symptomatic (12-14 weeks old) stage. Electrophysiological experiments were performed obtaining intracellular recordings in Mg2+ paralyzed phrenic nerve-hemidiaphragm preparations. Endplate potentials (EPPs), quantal content (q. c.) of EPPs, miniature endplate potentials (MEPPs) and giant miniature endplate potential (GMEPPs) were recorded. In the pre-symptomatic phase of the disease (4-6 weeks old mice), the selective A2A receptor agonist, CGS 21680, significantly enhanced (p<0.05 Unpaired t-test) the mean amplitude and q.c. of EPPs, and the frequency of MEPPs and GMEPPs at SOD1(G93A) neuromuscular junctions, the effect being of higher magnitude (p<0.05, Unpaired t-test) than age-matched control littermates. On the contrary, in symptomatic mice (12-14 weeks old), CGS 21680 was devoid of effect on both the amplitude and q.c. of EPPs and the frequency of MEPPs and GMEPPs (p<0.05 Paired t-test). The results herein reported clearly document that at the neuromuscular junction of SOD1(G93A) mice there is an exacerbation of A2A receptor-mediated excitatory effects at the pre-symptomatic phase, whereas in the symptomatic phase A2A receptor activation is absent. The results thus suggest that A2A receptors function changes with ALS progression.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/fisiopatologia , Mutação/genética , Junção Neuromuscular/fisiopatologia , Receptor A2A de Adenosina/metabolismo , Superóxido Dismutase/genética , Transmissão Sináptica , Adenosina/análogos & derivados , Esclerose Lateral Amiotrófica/patologia , Animais , Progressão da Doença , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Humanos , Masculino , Camundongos Transgênicos , Placa Motora/efeitos dos fármacos , Placa Motora/patologia , Placa Motora/fisiopatologia , Junção Neuromuscular/patologia , Fenetilaminas , Transmissão Sináptica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA