Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurosci Biobehav Rev ; 157: 105526, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38176632

RESUMEN

Schizophrenia is a complex disease related to combination and interactions between genetic and environmental factors, with an epigenetic influence. After the development of the first mono-factorial animal models of schizophrenia (1-hit), that reproduced patterns of either positive, negative and/or cognitive symptoms, more complex models combining two factors (2-hit) have been developed to better fit with the multifactorial etiology of the disease. In the two past decades, a new way to design animal models of schizophrenia have emerged by adding a third hit (3-hit). This review aims to discuss the relevance of the risk factors chosen for the tuning of the 3-hit animal models, as well as the validities measurements and their contribution to schizophrenia understanding. We intended to establish a comprehensive overview to help in the choice of factors for the design of multiple-hit animal models of schizophrenia.


Asunto(s)
Esquizofrenia , Animales , Esquizofrenia/genética , Modelos Animales de Enfermedad , Factores de Riesgo
2.
Biomolecules ; 13(5)2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37238621

RESUMEN

The subtype 6 of the serotoninergic receptors (5-HT6Rs) is highly expressed in the hippocampus, and evidence indicates the beneficial effects of 5-HT6Rs blockade on short- and long-term memory in rodents. Nevertheless, the underlying functional mechanisms still need to be established. To this end, we performed electrophysiological extracellular recordings to assess the effects of the 5-HT6Rs antagonist SB-271046 on the synaptic activity and functional plasticity at the CA3/CA1 hippocampal connections of male and female mice slices. We found that basal excitatory synaptic transmission and isolated N-methyl-D-aspartate receptors (NMDARs) activation were significantly increased by SB-271046. The NMDARs-related improvement was prevented by the GABAAR antagonist bicuculline in male but not in female mice. Regarding synaptic plasticity, neither paired-pulse facilitation (PPF) nor NMDARs-dependent long-term potentiation (LTP) (induced either by high-frequency or theta-burst stimulation) was affected by the 5-HT6Rs blockade. Taken together, our results indicate a sex-dependent 5-HT6Rs effect on synaptic activity at the CA3/CA1 hippocampal connections through changes in the excitation/inhibition balance.


Asunto(s)
Hipocampo , Sulfonamidas , Masculino , Femenino , Ratones , Animales , Hipocampo/metabolismo , Sulfonamidas/farmacología , Tiofenos/farmacología , Transmisión Sináptica , Receptores de N-Metil-D-Aspartato/metabolismo , Ácido gamma-Aminobutírico/farmacología
3.
Int J Mol Sci ; 24(5)2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36901735

RESUMEN

For almost half a century, acute hippocampal slice preparations have been widely used to investigate anti-amnesic (or promnesic) properties of drug candidates on long-term potentiation (LTP)-a cellular substrate that supports some forms of learning and memory. The large variety of transgenic mice models now available makes the choice of the genetic background when designing experiments crucially important. Furthermore, different behavioral phenotypes were reported between inbred and outbred strains. Notably, some differences in memory performance were emphasized. Despite this, investigations, unfortunately, did not explore electrophysiological properties. In this study, two stimulation paradigms were used to compare LTP in the hippocampal CA1 area of both inbred (C57BL/6) and outbred (NMRI) mice. High-frequency stimulation (HFS) revealed no strain difference, whereas theta-burst stimulation (TBS) resulted in significantly reduced LTP magnitude in NMRI mice. Additionally, we demonstrated that this reduced LTP magnitude (exhibited by NMRI mice) was due to lower responsiveness to theta-frequency during conditioning stimuli. In this paper, we discuss the anatomo-functional correlates that may explain such hippocampal synaptic plasticity divergence, although straightforward evidence is still lacking. Overall, our results support the prime importance of considering the animal model related to the intended electrophysiological experiments and the scientific issues to be addressed.


Asunto(s)
Hipocampo , Plasticidad Neuronal , Ratones , Animales , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Aprendizaje/fisiología , Ratones Endogámicos , Ratones Transgénicos , Estimulación Eléctrica
4.
Int J Mol Sci ; 23(24)2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36555191

RESUMEN

Impaired activation of the N-methyl-D-aspartate subtype of glutamate receptors (NMDAR) by D-serine is linked to cognitive aging. Whether this deregulation may be used to initiate pharmacological strategies has yet to be considered. To this end, we performed electrophysiological extracellular recordings at CA3/CA1 synapses in hippocampal slices from young and aged mice. We show that 0.1 nM of the soluble N-terminal recombinant fragment of the secreted amyloid-protein precursor-α (sAPPα) added in the bath significantly increased NMDAR activation in aged but not adult mice without impacting basal synaptic transmission. In addition, sAPPα rescued the age-related deficit of theta-burst-induced long-term potentiation. Significant NMDAR improvement occurred in adult mice when sAPPα was raised to 1 nM, and this effect was drastically reduced in transgenic mice deprived of D-serine through genetic deletion of the synthesizing enzyme serine racemase. Altogether, these results emphasize the interest to consider sAPPα treatment targeting D-serine-dependent NMDAR deregulation to alleviate cognitive aging.


Asunto(s)
Envejecimiento Saludable , Serina , Ratones , Animales , Serina/farmacología , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Precursor de Proteína beta-Amiloide/genética , Hipocampo/metabolismo , Sinapsis/metabolismo , Ratones Transgénicos
5.
Int J Mol Sci ; 22(5)2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33807989

RESUMEN

For a better translation from treatment designs of schizophrenia to clinical efficiency, there is a crucial need to refine preclinical animal models. In order to consider the multifactorial nature of the disorder, a new mouse model associating three factors (genetic susceptibility-partial deletion of the MAP6 gene, early-life stress-maternal separation, and pharmacological treatment-chronic Δ-9-tetrahydrocannabinol during adolescence) has recently been described. While this model depicts a schizophrenia-like phenotype, the neurobiological correlates remain unknown. Synaptic transmission and functional plasticity of the CA1 hippocampal region of male and female 3-hit mice were therefore investigated using electrophysiological recordings on the hippocampus slice. While basal excitatory transmission remained unaffected, NMDA receptor (NMDAr)-mediated long-term potentiation (LTP) triggered by theta-burst (TBS) but not by high-frequency (HFS) stimulation was impaired in 3-hit mice. Isolated NMDAr activation was not affected or even increased in female 3-hit mice, revealing a sexual dimorphism. Considering that the regulation of LTP is more prone to inhibitory tone if triggered by TBS than by HFS, the weaker potentiation in 3-hit mice suggests a deficiency of intrinsic GABA regulatory mechanisms. Indeed, NMDAr activation was increased by GABAA receptor blockade in wild-type but not in 3-hit mice. This electrophysiological study highlights dysregulations of functional properties and plasticity in hippocampal networks of 3-hit mice, one of the mechanisms suspected to contribute to the pathophysiology of schizophrenia. It also shows differences between males and females, supporting the sexual dimorphism observed in the disorder. Combined with the previously reported study, the present data reinforce the face validity of the 3-hit model that will help to consider new therapeutic strategies for psychosis.


Asunto(s)
Región CA1 Hipocampal/fisiopatología , Potenciación a Largo Plazo , Esquizofrenia/fisiopatología , Transmisión Sináptica , Ritmo Teta , Animales , Región CA1 Hipocampal/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Mutantes , Esquizofrenia/genética , Esquizofrenia/patología
6.
Cereb Cortex ; 31(1): 694-701, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32935845

RESUMEN

The type 4 serotonin receptor (5-HT4R) is highly involved in cognitive processes such as learning and memory. Behavioral studies have shown a beneficial effect of its activation and conversely reported memory impairments by its blockade. However, how modulation of 5HT4R enables modifications of hippocampal synaptic plasticity remains elusive. To shed light on the mechanisms at work, we investigated the effects of the 5-HT4R agonist RS67333 on long-term potentiation (LTP) within the hippocampal CA1 area. Although high-frequency stimulation-induced LTP remained unaffected by RS67333, the magnitude of LTP induced by theta-burst stimulation was significantly decreased. This effect was blocked by the selective 5-HT4R antagonist RS39604. Further, 5-HT4R-induced decrease in LTP magnitude was fully abolished in the presence of bicuculline, a GABAAR antagonist; hence, demonstrating involvement of GABA neurotransmission. In addition, we showed that the application of a GABABR antagonist, CGP55845, mimicked the effect of 5-HT4R activation, whereas concurrent application of CGP55845 and RS67333 did not elicit an additive inhibition effect on LTP. To conclude, through investigation of theta burst induced functional plasticity, we demonstrated an interplay between 5-HT4R activation and GABAergic neurotransmission within the hippocampal CA1 area.


Asunto(s)
Región CA1 Hipocampal/fisiología , Potenciación a Largo Plazo/fisiología , Plasticidad Neuronal/fisiología , Receptores de Serotonina 5-HT4/metabolismo , Animales , Estimulación Eléctrica/métodos , Potenciales Postsinápticos Excitadores/fisiología , Hipocampo/fisiología , Masculino , Ratones
7.
Int J Mol Sci ; 21(24)2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33322577

RESUMEN

d-serine is the major co-agonist of N-methyl-D-aspartate receptors (NMDAR) at CA3/CA1 hippocampal synapses, the activation of which drives long-term potentiation (LTP). The use of mice with targeted deletion of the serine racemase (SR) enzyme has been an important tool to uncover the physiological and pathological roles of D-serine. To date, some uncertainties remain regarding the direction of LTP changes in SR-knockout (SR-KO) mice, possibly reflecting differences in inhibitory GABAergic tone in the experimental paradigms used in the different studies. On the one hand, our extracellular recordings in hippocampal slices show that neither isolated NMDAR synaptic potentials nor LTP were altered in SR-KO mice. This was associated with a compensatory increase in hippocampal levels of glycine, another physiologic NMDAR co-agonist. SR-KO mice displayed no deficits in spatial learning, reference memory and cognitive flexibility. On the other hand, SR-KO mice showed a weaker LTP and a lower increase in NMDAR potentials compared to controls when GABAA receptors were pharmacologically blocked. Our results indicate that depletion of endogenous D-serine caused a reduced inhibitory activity in CA1 hippocampal networks, altering the excitatory/inhibitory balance, which contributes to preserve functional plasticity at synapses and to maintain related cognitive abilities.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Racemasas y Epimerasas/metabolismo , Aminoácidos/metabolismo , Animales , Electrofisiología , Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Masculino , Memoria/fisiología , Ratones , Prueba del Laberinto Acuático de Morris , Plasticidad Neuronal/fisiología , Racemasas y Epimerasas/genética , Receptores de N-Metil-D-Aspartato/metabolismo
8.
Brain ; 143(10): 2911-2928, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33103737

RESUMEN

Human post-natal neurodevelopmental delay is often associated with cerebral alterations that can lead, by themselves or associated with peripheral deficits, to premature death. Here, we report the clinical features of 10 patients from six independent families with mutations in the autosomal YIF1B gene encoding a ubiquitous protein involved in anterograde traffic from the endoplasmic reticulum to the cell membrane, and in Golgi apparatus morphology. The patients displayed global developmental delay, motor delay, visual deficits with brain MRI evidence of ventricle enlargement, myelination alterations and cerebellar atrophy. A similar profile was observed in the Yif1b knockout (KO) mouse model developed to identify the cellular alterations involved in the clinical defects. In the CNS, mice lacking Yif1b displayed neuronal reduction, altered myelination of the motor cortex, cerebellar atrophy, enlargement of the ventricles, and subcellular alterations of endoplasmic reticulum and Golgi apparatus compartments. Remarkably, although YIF1B was not detected in primary cilia, biallelic YIF1B mutations caused primary cilia abnormalities in skin fibroblasts from both patients and Yif1b-KO mice, and in ciliary architectural components in the Yif1b-KO brain. Consequently, our findings identify YIF1B as an essential gene in early post-natal development in human, and provide a new genetic target that should be tested in patients developing a neurodevelopmental delay during the first year of life. Thus, our work is the first description of a functional deficit linking Golgipathies and ciliopathies, diseases so far associated exclusively to mutations in genes coding for proteins expressed within the primary cilium or related ultrastructures. We therefore propose that these pathologies should be considered as belonging to a larger class of neurodevelopmental diseases depending on proteins involved in the trafficking of proteins towards specific cell membrane compartments.


Asunto(s)
Cilios/genética , Aparato de Golgi/genética , Mutación/genética , Trastornos del Neurodesarrollo/genética , Proteínas de Transporte Vesicular/genética , Animales , Células Cultivadas , Cilios/patología , Femenino , Aparato de Golgi/patología , Humanos , Masculino , Ratones , Ratones Noqueados , Trastornos del Neurodesarrollo/diagnóstico por imagen
9.
J Neurosci ; 40(34): 6489-6502, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32661027

RESUMEN

D-serine is a physiologic coagonist of NMDA receptors (NMDARs) required for synaptic plasticity, but mechanisms that terminate D-serine signaling are unclear. In particular, the identity of unidirectional plasma membrane transporters that mediate D-serine reuptake has remained elusive. We report that D-serine and glutamine share the same neuronal transport system, consisting of the classic system A transporters Slc38a1 and Slc38a2. We show that these transporters are not saturated with glutamine in vivo and regulate the extracellular levels of D-serine and NMDAR activity. Glutamine increased the NMDAR-dependent long-term potentiation and the isolated NMDAR potentials at the Schaffer collateral-CA1 synapses, but without affecting basal neurotransmission in male mice. Glutamine did not increase the NMDAR potentials in slices from serine racemase knock-out mice, which are devoid of D-serine, indicating that the effect of glutamine is caused by outcompeting D-serine for a dual glutamine-D-serine transport system. Inhibition of the system A reduced the uptake of D-serine in synaptosomes and neuronal cultures of mice of either sex, while increasing the extracellular D-serine concentration in slices and in vivo by microdialysis. When compared with Slc38a2, the Slc38a1 transporter displayed more favorable kinetics toward the D-enantiomer. Biochemical experiments with synaptosomes from Slc38a1 knock-down mice of either sex further support its role as a D-serine reuptake system. Our study identifies the first concentrative and electrogenic transporters mediating D-serine reuptake in vivo In addition to their classical role in the glutamine-glutamate cycle, system A transporters regulate the synaptic turnover of D-serine and its effects on NMDAR synaptic plasticity.SIGNIFICANCE STATEMENT Despite the plethora of roles attributed to D-serine, the regulation of its synaptic turnover is poorly understood. We identified the system A transporters Slc38a1 and Slc38a2 as the main pathway for neuronal reuptake of D-serine. These transporters are not saturated with glutamine in vivo and provide an unexpected link between the serine shuttle pathway, responsible for regulating D-serine synaptic turnover, and the glutamine-glutamate cycle. Our observations suggest that Slc38a1 and Slc38a2 have a dual role in regulating neurotransmission. In addition to their classical role as the glutamine providers, the system A transporters regulate extracellular D-serine and therefore affect NMDAR-dependent synaptic plasticity. Higher glutamine export from astrocytes would increase extracellular D-serine, providing a feedforward mechanism to increase synaptic NMDAR activation.


Asunto(s)
Sistema de Transporte de Aminoácidos A/metabolismo , Glutamina/metabolismo , Plasticidad Neuronal , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Transducción de Señal , Animales , Femenino , Hipocampo/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Transmisión Sináptica
10.
Sci Rep ; 9(1): 20138, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882891

RESUMEN

The prevalence of cognitive decline is increasing as the ageing population is considerably growing. Restricting this age-associated process has become a challenging public health issue. The age-related increase in oxidative stress plays a major role in cognitive decline, because of its harmful effect on functional plasticity of the brain, such as long-term potentiation (LTP). Here, we show that citrulline (Cit) has powerful antioxidant properties that can limit ex vivo oxidative stress-induced LTP impairment in the hippocampus. We also illustrate that a three-month Cit supplementation has a protective effect on LTP in aged rats in vivo. The identification of a Cit oxidation byproduct in vitro suggests that the antioxidant properties of Cit could result from its own oxidation. Cit supplementation may be a promising preventive nutritional approach to limit age-related cognitive decline.


Asunto(s)
Envejecimiento , Citrulina/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Envejecimiento/metabolismo , Animales , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatología , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Ratones , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas
11.
Proc Natl Acad Sci U S A ; 116(41): 20736-20742, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31548413

RESUMEN

Astrocytes express the 3-phosphoglycerate dehydrogenase (Phgdh) enzyme required for the synthesis of l-serine from glucose. Astrocytic l-serine was proposed to regulate NMDAR activity by shuttling to neurons to sustain d-serine production, but this hypothesis remains untested. We now report that inhibition of astrocytic Phgdh suppressed the de novo synthesis of l-and d-serine and reduced the NMDAR synaptic potentials and long-term potentiation (LTP) at the Schaffer collaterals-CA1 synapse. Likewise, enzymatic removal of extracellular l-serine impaired LTP, supporting an l-serine shuttle mechanism between glia and neurons in generating the NMDAR coagonist d-serine. Moreover, deletion of serine racemase (SR) in glutamatergic neurons abrogated d-serine synthesis to the same extent as Phgdh inhibition, suggesting that neurons are the predominant source of the newly synthesized d-serine. We also found that the synaptic NMDAR activation in adult SR-knockout (KO) mice requires Phgdh-derived glycine, despite the sharp decline in the postnatal glycine levels as a result of the emergence of the glycine cleavage system. Unexpectedly, we also discovered that glycine regulates d-serine metabolism by a dual mechanism. The first consists of tonic inhibition of SR by intracellular glycine observed in vitro, primary cultures, and in vivo microdialysis. The second involves a transient glycine-induce d-serine release through the Asc-1 transporter, an effect abolished in Asc-1 KO mice and diminished by deleting SR in glutamatergic neurons. Our observations suggest that glycine is a multifaceted regulator of d-serine metabolism and implicate both d-serine and glycine in mediating NMDAR synaptic activation at the mature hippocampus through a Phgdh-dependent shuttle mechanism.


Asunto(s)
Astrocitos/metabolismo , Glicina/metabolismo , Fosfoglicerato-Deshidrogenasa/metabolismo , Racemasas y Epimerasas/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Sinapsis/fisiología , Animales , Astrocitos/citología , Hipocampo/citología , Hipocampo/metabolismo , Potenciación a Largo Plazo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/metabolismo , Fosfoglicerato-Deshidrogenasa/genética , Receptores de N-Metil-D-Aspartato/genética
12.
Acta Neuropathol Commun ; 7(1): 46, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30885273

RESUMEN

Recent evidences suggest the involvement of DYRK1A (dual specificity tyrosine phosphorylation-regulated kinase 1 A) in Alzheimer's disease (AD). Here we showed that DYRK1A undergoes a proteolytic processing in AD patients hippocampus without consequences on its kinase activity. Resulting truncated forms accumulate in astrocytes and exhibit increased affinity towards STAT3ɑ, a regulator of inflammatory process. These findings were confirmed in APP/PS1 mice, an amyloid model of AD, suggesting that this DYRK1A cleavage is a consequence of the amyloid pathology. We identified in vitro the Leucettine L41 as a compound able to prevent DYRK1A proteolysis in both human and mouse protein extracts. We then showed that intraperitoneal injections of L41 in aged APP/PS1 mice inhibit STAT3ɑ phosphorylation and reduce pro-inflammatory cytokines levels (IL1- ß, TNF-ɑ and IL-12) associated to an increased microglial recruitment around amyloid plaques and decreased amyloid-ß plaque burden. Importantly, L41 treatment improved synaptic plasticity and rescued memory functions in APP/PS1 mice. Collectively, our results suggest that DYRK1A may contribute to AD pathology through its proteolytic process, reducing its kinase specificity. Further evaluation of inhibitors of DYRK1A truncation promises a new therapeutic approach for AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Fenotipo , Presenilina-1/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteolisis , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Animales , Hipocampo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Quinasas DyrK
13.
Acta Physiol (Oxf) ; 226(1): e13257, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30650253

RESUMEN

The last two decades have witnessed remarkable advance in our understanding the role of d-amino acids in the mammalian nervous system: from the unknown, to known molecules with unknown functions, to potential central players in health and disease. d-Amino acids have emerged as an important class of signaling molecules. In particular, the exploration of the roles of d-serine in brain physiopathology is a vibrant field that is growing at an accelerating pace. However, disentangling the functions of a chiral molecule in a complex chemical matrice as the brain requires specific measurement and detection methods but is also a challenging task as many molecular tools and models investigators are using can lead to confounded observations. Thus, study of d-amino acids demands accurate methodologies and specific controls, and these have often been lacking. Here we outline best practices for d-amino acid research, with a special emphasis on d-serine. We hope these concepts help move the field to greater rigor and reproducibility, allowing the field to advance.


Asunto(s)
Aminoácidos/metabolismo , Encéfalo/metabolismo , Serina/metabolismo , Animales , Humanos , Transmisión Sináptica/fisiología
14.
Front Mol Biosci ; 5: 106, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555832

RESUMEN

The N-methyl-D-Aspartate glutamate receptors (NMDARs) are pivotal for the functional and morphological plasticity that are required in neuronal networks for efficient brain activities and notably for cognitive-related abilities. Because NMDARs are heterogeneous in subunit composition and associated with multiple functional regulatory sites, their efficacy is under the tonic influence of numerous allosteric modulations, whose dysfunction generally represents the first step generating pathological states. Among the enzymatic candidates, serine racemase (SR) has recently gathered an increasing interest considering that it tightly regulates the production of d-serine, an amino acid now viewed as the main endogenous co-agonist necessary for NMDAR activation. Nowadays, SR deregulation is associated with a wide range of neurological and psychiatric diseases including schizophrenia, amyotrophic lateral sclerosis, and depression. This review aims at compelling the most recent experimental evidences indicating that changes in SR-related modulation of NMDARs also govern opposite functional dysfunctions in physiological and pathological (Alzheimer's disease) aging that finally results in memory disabilities in both cases. It also highlights SR as a relevant alternative target for new pharmacological strategies aimed at preventing functional alterations and cognitive impairments linked to the aging process.

15.
J Neurochem ; 147(4): 514-525, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30187927

RESUMEN

Activation of the N-methyl-D-aspartate subtype of glutamate receptor (NMDA-R) represents a key functional process for memory formation. A decreased synthesis of the NMDA-R co-agonist d-serine was recently proposed to contribute to alterations of hippocampus-dependent memory mechanisms with ageing. Nevertheless, other pathways could also be involved and thus considered to be targets of interest to prevent cognitive ageing. Herein, we demonstrate that the Asc-1 subtype of neutral amino acid (nAA) transporters that regulates d-serine and glycine release from neurons could be viewed as one of these targets. At CA3/CA1 hippocampal synapses, Asc-1 activation did not modify basal glutamate neurotransmission either in adult or aged rats. In contrast, Asc-1 activation significantly increased NMDA-R-dependent long-term potentiation (LTP) in both groups of animals and fully rescued the age-related LTP deficits. This rescue in aged animals was observed only when Asc-1 activation was selectively managed by d-Isoleucine (d-Ile), but not when less specifically driven by a mixture of nAA. Similarly, while any activation of Asc-1 improved the isolated NMDA-R-induced synaptic potentials in adult rats, only d-Ile was efficient in aged animals. Taken together, these results strengthen the interest in specifically targeting Asc-1 transporters to better cure age-associated memory decline. OPEN PRACTICES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Región CA1 Hipocampal/crecimiento & desarrollo , Región CA3 Hipocampal/crecimiento & desarrollo , Plasticidad Neuronal/fisiología , Envejecimiento/fisiología , Animales , Región CA1 Hipocampal/fisiología , Región CA3 Hipocampal/fisiología , Fenómenos Electrofisiológicos , Glicina/metabolismo , Potenciación a Largo Plazo , Masculino , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Transmisión Sináptica/fisiología
16.
Proc Natl Acad Sci U S A ; 115(38): 9628-9633, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30185558

RESUMEN

d-serine is a physiologic coagonist of NMDA receptors, but little is known about the regulation of its synthesis and synaptic turnover. The amino acid exchangers ASCT1 (Slc1a4) and ASCT2 (Slc1a5) are candidates for regulating d-serine levels. Using ASCT1 and ASCT2 KO mice, we report that ASCT1, rather than ASCT2, is a physiologic regulator of d-serine metabolism. ASCT1 is a major d-serine uptake system in astrocytes and can also export l-serine via heteroexchange, supplying neurons with the substrate for d-serine synthesis. ASCT1-KO mice display lower levels of brain d-serine along with higher levels of l-alanine, l-threonine, and glycine. Deletion of ASCT1 was associated with neurodevelopmental alterations including lower hippocampal and striatal volumes and changes in the expression of neurodevelopmental-relevant genes. Furthermore, ASCT1-KO mice exhibited deficits in motor function, spatial learning, and affective behavior, along with changes in the relative contributions of d-serine vs. glycine in mediating NMDA receptor activity. In vivo microdialysis demonstrated lower levels of extracellular d-serine in ASCT1-KO mice, confirming altered d-serine metabolism. These alterations are reminiscent of some of the neurodevelopmental phenotypes exhibited by patients with ASCT1 mutations. ASCT1-KO mice provide a useful model for potential therapeutic interventions aimed at correcting the metabolic impairments in patients with ASCT1 mutations.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Encéfalo/fisiología , Comunicación Celular/fisiología , Microcefalia/genética , Serina/metabolismo , Sistema de Transporte de Aminoácidos ASC/genética , Animales , Astrocitos/fisiología , Encéfalo/citología , Encéfalo/diagnóstico por imagen , Encéfalo/embriología , Modelos Animales de Enfermedad , Glicina/metabolismo , Células HEK293 , Humanos , Potenciación a Largo Plazo/fisiología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microcefalia/diagnóstico por imagen , Microcefalia/metabolismo , Microcefalia/patología , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/metabolismo , Neuronas/fisiología , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología
17.
Cereb Cortex ; 28(11): 3976-3993, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29048465

RESUMEN

The treatment of Alzheimer's disease (AD) remains challenging and requires a better in depth understanding of AD progression. Particularly, the link between amyloid protein precursor (APP) processing and Tau pathology development remains poorly understood. Growing evidences suggest that APP processing and amyloid-ß (Aß) release are upstream of Tau pathology but the lack of animal models mimicking the slow progression of human AD raised questions around this mechanism. Here, we described that an AD-like ßAPP processing in adults wild-type rats, yielding to human APP, ßCTF and Aß levels similar to those observed in AD patients, is sufficient to trigger gradual Tauopathy. The Tau hyperphosphorylation begins several months before the formation of both amyloid plaques and tangle-like aggregates in aged rats and without associated inflammation. Based on a longitudinal characterization over 30 months, we showed that extrasynaptic and emotional impairments appear before long-term potentiation deficits and memory decline and so before Aß and Tau aggregations. These compelling data allowed us to (1) experimentally confirm the causal relationship between ßAPP processing and Tau pathology in vivo and without Tau transgene overexpression, (2) support the amyloidogenic cascade and (3) propose a 4-step hypothesis of prodromal AD progression.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Péptidos beta-Amiloides/metabolismo , Animales , Progresión de la Enfermedad , Femenino , Vectores Genéticos , Humanos , Potenciación a Largo Plazo , Masculino , Fragmentos de Péptidos/metabolismo , Placa Amiloide/metabolismo , Presenilina-1/genética , Agregación Patológica de Proteínas/metabolismo , Ratas Wistar
18.
Cereb Cortex ; 27(2): 1573-1587, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26796213

RESUMEN

d-Serine is a co-agonist of NMDA receptors (NMDARs) whose activity is potentially regulated by Asc-1 (SLC7A10), a transporter that displays high affinity for d-serine and glycine. Asc-1 operates as a facilitative transporter and as an antiporter, though the preferred direction of d-serine transport is uncertain. We developed a selective Asc-1 blocker, Lu AE00527, that blocks d-serine release mediated by all the transport modes of Asc-1 in primary cultures and neocortical slices. Furthermore, d-serine release is reduced in slices from Asc-1 knockout (KO) mice, indicating that d-serine efflux is the preferred direction of Asc-1. The selectivity of Lu AE00527 is assured by the lack of effect on slices from Asc-1-KO mice, and the lack of interaction with the co-agonist site of NMDARs. Moreover, in vivo injection of Lu AE00527 in P-glycoprotein-deficient mice recapitulates a hyperekplexia-like phenotype similar to that in Asc-1-KO mice. In slices, Lu AE00527 decreases the long-term potentiation at the Schaffer collateral-CA1 synapses, but does not affect the long-term depression. Lu AE00527 blocks NMDAR synaptic potentials when typical Asc-1 extracellular substrates are present, but it does not affect AMPAR transmission. Our data demonstrate that Asc-1 mediates tonic co-agonist release, which is required for optimal NMDAR activation and synaptic plasticity.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/genética , Potenciación a Largo Plazo/fisiología , Plasticidad Neuronal/fisiología , Prosencéfalo/fisiología , Sinapsis/fisiología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Humanos , Ratones Noqueados , Neuronas/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología
19.
J Cereb Blood Flow Metab ; 37(2): 396-412, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26762506

RESUMEN

Insulin-like growth factors control numerous processes, namely somatic growth, metabolism and stress resistance, connecting this pathway to aging and age-related diseases. Insulin-like growth factor signaling also impacts on neurogenesis, neuronal survival and structural plasticity. Recent reports demonstrated that diminished insulin-like growth factor signaling confers increased stress resistance in brain and other tissues. To better understand the role of neuronal insulin-like growth factor signaling in neuroprotection, we inactivated insulin-like growth factor type-1-receptor in forebrain neurons using conditional Cre-LoxP-mediated gene targeting. We found that brain structure and function, including memory performance, were preserved in insulin-like growth factor receptor mutants, and that certain characteristics improved, notably synaptic transmission in hippocampal neurons. To reveal stress-related roles of insulin-like growth factor signaling, we challenged the brain using a stroke-like insult. Importantly, when charged with hypoxia-ischemia, mutant brains were broadly protected from cell damage, neuroinflammation and cerebral edema. We also found that in mice with insulin-like growth factor receptor knockout specifically in forebrain neurons, a substantial systemic upregulation of growth hormone and insulin-like growth factor-I occurred, which was associated with significant somatic overgrowth. Collectively, we found strong evidence that blocking neuronal insulin-like growth factor signaling increases peripheral somatotropic tone and simultaneously protects the brain against hypoxic-ischemic injury, findings that may contribute to developing new therapeutic concepts preventing the disabling consequences of stroke.


Asunto(s)
Eliminación de Gen , Hormona del Crecimiento/metabolismo , Neuroprotección , Prosencéfalo/patología , Receptor IGF Tipo 1/genética , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Prosencéfalo/metabolismo , Accidente Cerebrovascular/metabolismo , Regulación hacia Arriba
20.
PLoS One ; 11(2): e0148328, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26849367

RESUMEN

We studied the role of testosterone, mediated by the androgen receptor (AR), in modulating temporal order memory for visual objects. For this purpose, we used male mice lacking AR specifically in the nervous system. Control and mutant males were gonadectomized at adulthood and supplemented with equivalent amounts of testosterone in order to normalize their hormonal levels. We found that neural AR deletion selectively impaired the processing of temporal information for visual objects, without affecting classical object recognition or anxiety-like behavior and circulating corticosterone levels, which remained similar to those in control males. Thus, mutant males were unable to discriminate between the most recently seen object and previously seen objects, whereas their control littermates showed more interest in exploring previously seen objects. Because the hippocampal CA1 area has been associated with temporal memory for visual objects, we investigated whether neural AR deletion altered the functionality of this region. Electrophysiological analysis showed that neural AR deletion affected basal glutamate synaptic transmission and decreased the magnitude of N-methyl-D-aspartate receptor (NMDAR) activation and high-frequency stimulation-induced long-term potentiation. The impairment of NMDAR function was not due to changes in protein levels of receptor. These results provide the first evidence for the modulation of temporal processing of information for visual objects by androgens, via AR activation, possibly through regulation of NMDAR signaling in the CA1 area in male mice.


Asunto(s)
Hipocampo/fisiología , Receptores Androgénicos/metabolismo , Procesamiento Espacial/fisiología , Animales , Ansiedad/genética , Conducta Animal , Corticosterona/sangre , Fenómenos Electrofisiológicos , Eliminación de Gen , Potenciación a Largo Plazo/fisiología , Masculino , Memoria a Corto Plazo , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal , Receptores Androgénicos/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...