Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Methods Cell Biol ; 188: 237-254, 2024.
Article in English | MEDLINE | ID: mdl-38880526

ABSTRACT

The prevalence of central nervous system (CNS) dysfunction as a result of disease or trauma remains a clinically unsolved problem which is raising increased awareness in our aging society. Human Dental Pulp Stem Cells (hDPSCs) are excellent candidates to be used in tissue engineering and regenerative therapies of the CNS due to their neural differentiation ability and lack of tumorigenicity. Accordingly, they have been successfully used in animal models of spinal cord injury, stroke and peripheral neuropathies. The ideal therapy in brain injury should combine strategies aiming to protect the damaged lesion and, at the same time, accelerate brain tissue regeneration, thus promoting fast recovery while minimizing side or long-term effects. The use of bioresorbable nanopatterned poly(lactide-co-ɛ-caprolactone) (PLCL) polymeric scaffolds as hDPCSs carriers can represent an advantage for tissue regeneration. In this chapter, we describe the surgical procedures to implant functionalized bioresorbable scaffolds loaded with hDPSCs to improve the brain lesion microenvironment in an intracranial stab wound injury model severing the rostral migratory stream (RMS) that connects the brain subventricular zone (SVZ) and the olfactory bulb in nude mice. Additionally, we also describe the technical steps after animal sacrifice for histological tissue observation and characterization.


Subject(s)
Dental Pulp , Disease Models, Animal , Mice, Nude , Stem Cells , Tissue Scaffolds , Dental Pulp/cytology , Animals , Humans , Tissue Scaffolds/chemistry , Mice , Stem Cells/cytology , Stem Cell Transplantation/methods , Wounds, Stab/therapy , Absorbable Implants , Brain Injuries/therapy , Brain Injuries/pathology , Tissue Engineering/methods
2.
EMBO J ; 43(4): 533-567, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38316990

ABSTRACT

The phospholipid and free fatty acid (FFA) composition of neuronal membranes plays a crucial role in learning and memory, but the mechanisms through which neuronal activity affects the brain's lipid landscape remain largely unexplored. The levels of saturated FFAs, particularly of myristic acid (C14:0), strongly increase during neuronal stimulation and memory acquisition, suggesting the involvement of phospholipase A1 (PLA1) activity in synaptic plasticity. Here, we show that genetic ablation of the PLA1 isoform DDHD2 in mice dramatically reduces saturated FFA responses to memory acquisition across the brain. Furthermore, DDHD2 loss also decreases memory performance in reward-based learning and spatial memory models prior to the development of neuromuscular deficits that mirror human spastic paraplegia. Via pulldown-mass spectrometry analyses, we find that DDHD2 binds to the key synaptic protein STXBP1. Using STXBP1/2 knockout neurosecretory cells and a haploinsufficient STXBP1+/- mouse model of human early infantile encephalopathy associated with intellectual disability and motor dysfunction, we show that STXBP1 controls targeting of DDHD2 to the plasma membrane and generation of saturated FFAs in the brain. These findings suggest key roles for DDHD2 and STXBP1 in lipid metabolism and in the processes of synaptic plasticity, learning, and memory.


Subject(s)
Fatty Acids, Nonesterified , Memory, Long-Term , Munc18 Proteins , Phospholipases , Animals , Mice , Brain/metabolism , Fatty Acids, Nonesterified/metabolism , Memory/physiology , Munc18 Proteins/genetics , Phospholipases/genetics
3.
Nat Commun ; 14(1): 8312, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38097535

ABSTRACT

The consolidation of recent memories depends on memory replays, also called ripples, generated within the hippocampus during slow-wave sleep, and whose inactivation leads to memory impairment. For now, the mobilisation, localisation and importance of synaptic plasticity events associated to ripples are largely unknown. To tackle this question, we used cell surface AMPAR immobilisation to block post-synaptic LTP within the hippocampal region of male mice during a spatial memory task, and show that: 1- hippocampal synaptic plasticity is engaged during consolidation, but is dispensable during encoding or retrieval. 2- Plasticity blockade during sleep results in apparent forgetting of the encoded rule. 3- In vivo ripple recordings show a strong effect of AMPAR immobilisation when a rule has been recently encoded. 4- In situ investigation suggests that plasticity at CA3-CA3 recurrent synapses supports ripple generation. We thus propose that post-synaptic AMPAR mobility at CA3 recurrent synapses is necessary for ripple-dependent rule consolidation.


Subject(s)
Memory Consolidation , Mice , Male , Animals , Memory Consolidation/physiology , Hippocampus/physiology , Neuronal Plasticity/physiology , Sleep/physiology , Spatial Memory , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology
4.
Front Cell Neurosci ; 17: 1212202, 2023.
Article in English | MEDLINE | ID: mdl-37435048

ABSTRACT

Imbalance between excitation and inhibition in the cerebral cortex is one of the main theories in neuropsychiatric disorder pathophysiology. Cortical inhibition is finely regulated by a variety of highly specialized GABAergic interneuron types, which are thought to organize neural network activities. Among interneurons, axo-axonic cells are unique in making synapses with the axon initial segment of pyramidal neurons. Alterations of axo-axonic cells have been proposed to be implicated in disorders including epilepsy, schizophrenia and autism spectrum disorder. However, evidence for the alteration of axo-axonic cells in disease has only been examined in narrative reviews. By performing a systematic review of studies investigating axo-axonic cells and axo-axonic communication in epilepsy, schizophrenia and autism spectrum disorder, we outline convergent findings and discrepancies in the literature. Overall, the implication of axo-axonic cells in neuropsychiatric disorders might have been overstated. Additional work is needed to assess initial, mostly indirect findings, and to unravel how defects in axo-axonic cells translates to cortical dysregulation and, in turn, to pathological states.

5.
Sci Adv ; 8(30): eabm5298, 2022 Jul 29.
Article in English | MEDLINE | ID: mdl-35895810

ABSTRACT

Regulation of synaptic neurotransmitter receptor content is a fundamental mechanism for tuning synaptic efficacy during experience-dependent plasticity and behavioral adaptation. However, experimental approaches to track and modify receptor movements in integrated experimental systems are limited. Exploiting AMPA-type glutamate receptors (AMPARs) as a model, we generated a knock-in mouse expressing the biotin acceptor peptide (AP) tag on the GluA2 extracellular N-terminal. Cell-specific introduction of biotin ligase allows the use of monovalent or tetravalent avidin variants to respectively monitor or manipulate the surface mobility of endogenous AMPAR containing biotinylated AP-GluA2 in neuronal subsets. AMPAR immobilization precluded the expression of long-term potentiation and formation of contextual fear memory, allowing target-specific control of the expression of synaptic plasticity and animal behavior. The AP tag knock-in model offers unprecedented access to resolve and control the spatiotemporal dynamics of endogenous receptors, and opens new avenues to study the molecular mechanisms of synaptic plasticity and learning.

6.
Nat Commun ; 12(1): 1557, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33692361

ABSTRACT

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.


Subject(s)
Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Fragile X Mental Retardation Protein/metabolism , Mutation, Missense/physiology , Receptors, Glutamate/metabolism , Animals , Biotinylation , Brain/metabolism , Brain/physiopathology , Cells, Cultured , Cognitive Dysfunction/metabolism , Female , Fragile X Mental Retardation Protein/genetics , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Immunoblotting , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Male , Mice , Mutation, Missense/genetics , Patch-Clamp Techniques , Receptors, Glutamate/genetics
7.
Elife ; 92020 11 30.
Article in English | MEDLINE | ID: mdl-33252331

ABSTRACT

Survival depends on the ability of animals to select the appropriate behavior in response to threat and safety sensory cues. However, the synaptic and circuit mechanisms by which the brain learns to encode accurate predictors of threat and safety remain largely unexplored. Here, we show that frontal association cortex (FrA) pyramidal neurons of mice integrate auditory cues and basolateral amygdala (BLA) inputs non-linearly in a NMDAR-dependent manner. We found that the response of FrA pyramidal neurons was more pronounced to Gaussian noise than to pure frequency tones, and that the activation of BLA-to-FrA axons was the strongest in between conditioning pairings. Blocking BLA-to-FrA signaling specifically at the time of presentation of Gaussian noise (but not 8 kHz tone) between conditioning trials impaired the formation of auditory fear memories. Taken together, our data reveal a circuit mechanism that facilitates the formation of fear traces in the FrA, thus providing a new framework for probing discriminative learning and related disorders.


Subject(s)
Acoustic Stimulation/adverse effects , Amygdala/physiology , Fear/physiology , Frontal Lobe/physiology , Learning/physiology , Animals , Calcium/metabolism , Conditioning, Classical/physiology , Male , Mice , Microscopy, Confocal , Neuronal Plasticity/physiology , Optogenetics , Patch-Clamp Techniques
8.
Elife ; 82019 10 30.
Article in English | MEDLINE | ID: mdl-31663854

ABSTRACT

Glutamate secretion at excitatory synapses is tightly regulated to allow for the precise tuning of synaptic strength. Vesicular Glutamate Transporters (VGLUT) accumulate glutamate into synaptic vesicles (SV) and thereby regulate quantal size. Further, the number of release sites and the release probability of SVs maybe regulated by the organization of active-zone proteins and SV clusters. In the present work, we uncover a mechanism mediating an increased SV clustering through the interaction of VGLUT1 second proline-rich domain, endophilinA1 and intersectin1. This strengthening of SV clusters results in a combined reduction of axonal SV super-pool size and miniature excitatory events frequency. Our findings support a model in which clustered vesicles are held together through multiple weak interactions between Src homology three and proline-rich domains of synaptic proteins. In mammals, VGLUT1 gained a proline-rich sequence that recruits endophilinA1 and turns the transporter into a regulator of SV organization and spontaneous release.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Glutamates/metabolism , Synaptic Vesicles/metabolism , Vesicular Glutamate Transport Protein 1/metabolism , Animals , Biological Transport , Humans , Mice , Mice, Knockout , Rats , Vesicular Glutamate Transport Protein 1/deficiency
9.
Nat Neurosci ; 22(10): 1536-1543, 2019 10.
Article in English | MEDLINE | ID: mdl-31477899

ABSTRACT

Activity-dependent synaptic plasticity has since long been proposed to represent the subcellular substrate of learning and memory, one of the most important behavioral processes through which we adapt to our environment. Despite the undisputed importance of synaptic plasticity for brain function, its exact contribution to learning processes in the context of cellular and connectivity modifications remains obscure. Causally bridging synaptic and behavioral modifications indeed remains limited by the available tools to measure and control synaptic strength and plasticity in vivo under behaviorally relevant conditions. After a brief summary of the current state of knowledge of the links between synaptic plasticity and learning, we will review and discuss the available and desired tools to progress in this endeavor.


Subject(s)
Learning/physiology , Neuronal Plasticity/physiology , Animals , Brain/cytology , Brain/physiology , Humans , Neural Pathways/cytology , Neural Pathways/physiology
10.
Front Cell Neurosci ; 13: 220, 2019.
Article in English | MEDLINE | ID: mdl-31164805

ABSTRACT

The amygdala, and more precisely its lateral nucleus, is thought to attribute emotional valence to external stimuli by generating long-term plasticity changes at long-range projections to principal cells. Aversive experience has also been shown to modify pre- and post-synaptic markers in the amygdala, suggesting their possible role in the structural organization of adult amygdala networks. Here, we focused on how the maturation of cortical and thalamic long-range projections occurs on principal neurons and interneurons in the lateral amygdala (LA). We performed dual electrophysiological recordings of identified cells in juvenile and adult GAD67-GFP mice after independent stimulation of cortical and thalamic afferent systems. The results demonstrate that synaptic strengthening occurs during development at synapses projecting to LA principal neurons, but not interneurons. As synaptic strengthening underlies fear conditioning which depends, in turn, on presence and increasing expression of synapsin I, we tested if synapsin I contributes to synaptic strengthening during development. Interestingly, the physiological synaptic strengthening of cortical and thalamic synapses projecting to LA principal neurons was virtually abolished in synapsin I knockout mice, but not differences were observed in the excitatory projections to interneurons. Immunohistochemistry analysis showed that the presence of synapsin I is restricted to excitatory contacts projecting to principal neurons in LA of adult mice. These results indicate that synapsin I is a key regulator of the maturation of synaptic connectivity in this brain region and that is expression is dependent on postsynaptic identity.

11.
Nat Commun ; 9(1): 4272, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30323233

ABSTRACT

Impaired hippocampal synaptic plasticity contributes to cognitive impairment in Huntington's disease (HD). However, the molecular basis of such synaptic plasticity defects is not fully understood. Combining live-cell nanoparticle tracking and super-resolution imaging, we show that AMPAR surface diffusion, a key player in synaptic plasticity, is disturbed in various rodent models of HD. We demonstrate that defects in the brain-derived neurotrophic factor (BDNF)-tyrosine receptor kinase B (TrkB) signaling pathway contribute to the deregulated AMPAR trafficking by reducing the interaction between transmembrane AMPA receptor regulatory proteins (TARPs) and the PDZ-domain scaffold protein PSD95. The disturbed AMPAR surface diffusion is rescued by the antidepressant drug tianeptine via the BDNF signaling pathway. Tianeptine also restores the impaired LTP and hippocampus-dependent memory in different HD mouse models. These findings unravel a mechanism underlying hippocampal synaptic and memory dysfunction in HD, and highlight AMPAR surface diffusion as a promising therapeutic target.


Subject(s)
Hippocampus/physiopathology , Huntington Disease/physiopathology , Memory/physiology , Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Diffusion , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Huntington Disease/metabolism , Huntington Disease/pathology , Long-Term Potentiation/drug effects , Memory/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/drug effects , Neuronal Plasticity/drug effects , Protein Transport/drug effects , Receptor, trkB/metabolism , Signal Transduction/drug effects , Synapses/drug effects , Synapses/metabolism , Thiazepines/pharmacology
12.
Hum Mol Genet ; 27(12): 2138-2153, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29659809

ABSTRACT

The aristaless-related homeobox (ARX) transcription factor is involved in the development of GABAergic and cholinergic neurons in the forebrain. ARX mutations have been associated with a wide spectrum of neurodevelopmental disorders in humans, among which the most frequent, a 24 bp duplication in the polyalanine tract 2 (c.428_451dup24), gives rise to intellectual disability, fine motor defects with or without epilepsy. To understand the functional consequences of this mutation, we generated a partially humanized mouse model carrying the c.428_451dup24 duplication (Arxdup24/0) that we characterized at the behavior, neurological and molecular level. Arxdup24/0 males presented with hyperactivity, enhanced stereotypies and altered contextual fear memory. In addition, Arxdup24/0 males had fine motor defects with alteration of reaching and grasping abilities. Transcriptome analysis of Arxdup24/0 forebrains at E15.5 showed a down-regulation of genes specific to interneurons and an up-regulation of genes normally not expressed in this cell type, suggesting abnormal interneuron development. Accordingly, interneuron migration was altered in the cortex and striatum between E15.5 and P0 with consequences in adults, illustrated by the defect in the inhibitory/excitatory balance in Arxdup24/0 basolateral amygdala. Altogether, we showed that the c.428_451dup24 mutation disrupts Arx function with a direct consequence on interneuron development, leading to hyperactivity and defects in precise motor movement control and associative memory. Interestingly, we highlighted striking similarities between the mouse phenotype and a cohort of 33 male patients with ARX c.428_451dup24, suggesting that this new mutant mouse line is a good model for understanding the pathophysiology and evaluation of treatment.


Subject(s)
Epilepsy/genetics , Homeodomain Proteins/genetics , Neurodevelopmental Disorders/genetics , Transcription Factors/genetics , Adolescent , Adult , Animals , Child , Child, Preschool , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Contracture , Disease Models, Animal , Epilepsy/physiopathology , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gene Expression Regulation, Developmental , Humans , Infant , Intellectual Disability , Male , Mice , Mutation , Neurodevelopmental Disorders/physiopathology , Peptides/genetics , Prosencephalon/physiopathology , Spastic Paraplegia, Hereditary , Transcriptome/genetics , Young Adult
13.
Prog Neuropsychopharmacol Biol Psychiatry ; 84(Pt B): 392-397, 2018 06 08.
Article in English | MEDLINE | ID: mdl-28774568

ABSTRACT

The amygdala is a part of the limbic circuit that has been extensively studied in terms of synaptic connectivity, plasticity and cellular organization since decades (Ehrlich et al., 2009; Ledoux, 2000; Maren, 2001). Amygdala sub-nuclei, including lateral, basolateral and central amygdala appear now as "hubs" providing in parallel and in series neuronal processing enabling the animal to elicit freezing or escaping behavior in response to external threats. In rodents, these behaviors are easily observed and quantified following associative fear conditioning. Thus, studies on amygdala circuit in association with threat/fear behavior became very popular in laboratories and are often used among other behavioral tests to evaluate learning abilities of mouse models for various neuropsychiatric conditions including genetically encoded intellectual disabilities (ID). Yet, more than 100 human X-linked genes - and several hundreds of autosomal genes - have been associated with ID in humans. These mutations introduced in mice can generate social deficits, anxiety dysregulations and fear learning impairments (McNaughton et al., 2008; Houbaert et al., 2013; Jayachandran et al., 2014; Zhang et al., 2015). Noteworthy, a significant proportion of the coded ID gene products are synaptic proteins. It is postulated that the loss of function of these proteins could destabilize neuronal circuits by global changes of the balance between inhibitory and excitatory drives onto neurons. However, whereas amygdala related behavioral deficits are commonly observed in ID models, the role of most of these ID-genes in synaptic function and plasticity in the amygdala are only sparsely studied. We will here discuss some of the concepts that emerged from amygdala-targeted studies examining the role of syndromic and non-syndromic ID genes in fear-related behaviors and/or synaptic function. Along describing these cases, we will discuss how synaptic deficits observed in amygdala circuits could impact memory formation and expression of conditioned fear.


Subject(s)
Amygdala/pathology , Intellectual Disability/pathology , Synapses/pathology , Animals , Avoidance Learning/physiology , Conditioning, Classical/physiology , Disease Models, Animal , Fear , Humans , Intellectual Disability/genetics , Intellectual Disability/physiopathology
14.
J Neurosci ; 37(46): 11114-11126, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29030432

ABSTRACT

Classical and systems genetics have identified wide networks of genes associated with cognitive and neurodevelopmental diseases. In parallel to deciphering the role of each of these genes in neuronal or synaptic function, evaluating the response of neuronal and molecular networks to gene loss of function could reveal some pathophysiological mechanisms potentially accessible to nongenetic therapies. Loss of function of the Rho-GAP oligophrenin-1 is associated with cognitive impairments in both human and mouse. Upregulation of both PKA and ROCK has been reported in Ophn1-/y mice, but it remains unclear whether kinase hyperactivity contributes to the behavioral phenotypes. In this study, we thoroughly characterized a prominent perseveration phenotype displayed by Ophn1-deficient mice using a Y-maze spatial working memory (SWM) test. We report that Ophn1 deficiency in the mouse generated severe cognitive impairments, characterized by both a high occurrence of perseverative behaviors and a lack of deliberation during the SWM test. In vivo and in vitro pharmacological experiments suggest that PKA dysregulation in the mPFC underlies cognitive dysfunction in Ophn1-deficient mice, as assessed using a delayed spatial alternation task results. Functionally, mPFC neuronal networks appeared to be affected in a PKA-dependent manner, whereas hippocampal-PFC projections involved in SWM were not affected in Ophn1-/y mice. Thus, we propose that discrete gene mutations in intellectual disability might generate "secondary" pathophysiological mechanisms, which are prone to become pharmacological targets for curative strategies in adult patients.SIGNIFICANCE STATEMENT Here we report that Ophn1 deficiency generates severe impairments in performance at spatial working memory tests, characterized by a high occurrence of perseverative behaviors and a lack of decision making. This cognitive deficit is consecutive to PKA deregulation in the mPFC that prevents Ophn1 KO mice to exploit a correctly acquired rule. Functionally, mPFC neuronal networks appear to be affected in a PKA-dependent manner, whereas behaviorally important hippocampal projections were preserved by the mutation. Thus, we propose that discrete gene mutations in intellectual disability can generate "secondary" pathophysiological mechanisms prone to become pharmacological targets for curative strategies in adults.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeletal Proteins/deficiency , GTPase-Activating Proteins/deficiency , Memory Disorders/metabolism , Memory, Short-Term/physiology , Nuclear Proteins/deficiency , Prefrontal Cortex/metabolism , Animals , Male , Maze Learning/physiology , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Net/metabolism , Nerve Net/physiopathology , Organ Culture Techniques , Prefrontal Cortex/physiopathology , Random Allocation
15.
PLoS Genet ; 13(7): e1006886, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28704368

ABSTRACT

Koolen-de Vries syndrome (KdVS) is a multi-system disorder characterized by intellectual disability, friendly behavior, and congenital malformations. The syndrome is caused either by microdeletions in the 17q21.31 chromosomal region or by variants in the KANSL1 gene. The reciprocal 17q21.31 microduplication syndrome is associated with psychomotor delay, and reduced social interaction. To investigate the pathophysiology of 17q21.31 microdeletion and microduplication syndromes, we generated three mouse models: 1) the deletion (Del/+); or 2) the reciprocal duplication (Dup/+) of the 17q21.31 syntenic region; and 3) a heterozygous Kansl1 (Kans1+/-) model. We found altered weight, general activity, social behaviors, object recognition, and fear conditioning memory associated with craniofacial and brain structural changes observed in both Del/+ and Dup/+ animals. By investigating hippocampus function, we showed synaptic transmission defects in Del/+ and Dup/+ mice. Mutant mice with a heterozygous loss-of-function mutation in Kansl1 displayed similar behavioral and anatomical phenotypes compared to Del/+ mice with the exception of sociability phenotypes. Genes controlling chromatin organization, synaptic transmission and neurogenesis were upregulated in the hippocampus of Del/+ and Kansl1+/- animals. Our results demonstrate the implication of KANSL1 in the manifestation of KdVS phenotypes and extend substantially our knowledge about biological processes affected by these mutations. Clear differences in social behavior and gene expression profiles between Del/+ and Kansl1+/- mice suggested potential roles of other genes affected by the 17q21.31 deletion. Together, these novel mouse models provide new genetic tools valuable for the development of therapeutic approaches.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Duplication/genetics , Cognition , Intellectual Disability/genetics , Nuclear Proteins/genetics , Animals , Body Weight , Brain/metabolism , Brain/ultrastructure , Chromosome Deletion , Chromosome Structures/genetics , Chromosome Structures/metabolism , Chromosomes, Human, Pair 17/genetics , DNA Copy Number Variations , Disease Models, Animal , Epigenesis, Genetic , Female , Gene Deletion , Gene Rearrangement , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/genetics , Nuclear Proteins/metabolism , Synaptic Transmission/genetics , Up-Regulation
16.
Hum Mol Genet ; 25(11): 2314-2323, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27146843

ABSTRACT

Loss of function mutations in human Oligophrenin1 (OPHN1) gene are responsible for syndromic intellectual disability (ID) associated with cerebellar hypoplasia and cerebral ventricles enlargement. Functional studies in rodent models suggest that OPHN1 linked ID is a consequence of abnormal synaptic transmission and shares common pathophysiological mechanisms with other cognitive disorders. Variants of this gene have been also identified in autism spectrum disorder and schizophrenia. The advanced understanding of the mechanisms underlying OPHN1-related ID, allowed us to develop a therapeutic approach targeting the Ras homolog gene family, member A (RHOA) signalling pathway and repurpose Fasudil- a well-tolerated Rho Kinase (ROCK) and Protein Kinase A (PKA) inhibitor- as a treatment of ID. We have previously shown ex-vivo its beneficial effect on synaptic transmission and plasticity in a mouse model of the OPHN1 loss of function. Here, we report that chronic treatment in adult mouse with Fasudil, is able to counteract vertical and horizontal hyperactivities, restores recognition memory and limits the brain ventricular dilatation observed in Ophn1-/y However, deficits in working and spatial memories are partially or not rescued by the treatment. These results highlight the potential of Fasudil treatment in synaptopathies and also the need for multiple therapeutic approaches especially in adult where brain plasticity is reduced.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Brain/physiopathology , Cytoskeletal Proteins/genetics , GTPase-Activating Proteins/genetics , Intellectual Disability/drug therapy , Nuclear Proteins/genetics , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/administration & dosage , Adult , Animals , Autism Spectrum Disorder , Behavior, Animal/drug effects , Brain/drug effects , Disease Models, Animal , Humans , Intellectual Disability/genetics , Intellectual Disability/physiopathology , Mice , Synaptic Transmission
17.
Mol Autism ; 7: 1, 2016.
Article in English | MEDLINE | ID: mdl-26753090

ABSTRACT

BACKGROUND: Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders caused by the interaction between genetic vulnerability and environmental factors. MicroRNAs (miRNAs) are key posttranscriptional regulators involved in multiple aspects of brain development and function. Previous studies have investigated miRNAs expression in ASD using non-neural cells like lymphoblastoid cell lines (LCL) or postmortem tissues. However, the relevance of LCLs is questionable in the context of a neurodevelopmental disorder, and the impact of the cause of death and/or post-death handling of tissue likely contributes to the variations observed between studies on brain samples. METHODS: miRNA profiling using TLDA high-throughput real-time qPCR was performed on miRNAs extracted from olfactory mucosal stem cells (OMSCs) biopsied from eight patients and six controls. This tissue is considered as a closer tissue to neural stem cells that could be sampled in living patients and was never investigated for such a purpose before. Real-time PCR was used to validate a set of differentially expressed miRNAs, and bioinformatics analysis determined common pathways and gene targets. Luciferase assays and real-time PCR analysis were used to evaluate the effect of miRNAs misregulation on the expression and translation of several autism-related transcripts. Viral vector-mediated expression was used to evaluate the impact of miRNAs deregulation on neuronal or glial cells functions. RESULTS: We identified a signature of four miRNAs (miR-146a, miR-221, miR-654-5p, and miR-656) commonly deregulated in ASD. This signature is conserved in primary skin fibroblasts and may allow discriminating between ASD and intellectual disability samples. Putative target genes of the differentially expressed miRNAs were enriched for pathways previously associated to ASD, and altered levels of neuronal transcripts targeted by miR-146a, miR-221, and miR-656 were observed in patients' cells. In the mouse brain, miR-146a, and miR-221 display strong neuronal expression in regions important for high cognitive functions, and we demonstrated that reproducing abnormal miR-146a expression in mouse primary cell cultures leads to impaired neuronal dendritic arborization and increased astrocyte glutamate uptake capacities. CONCLUSIONS: While independent replication experiments are needed to clarify whether these four miRNAS could serve as early biomarkers of ASD, these findings may have important diagnostic implications. They also provide mechanistic connection between miRNA dysregulation and ASD pathophysiology and may open up new opportunities for therapeutic.


Subject(s)
Adult Stem Cells/metabolism , Autism Spectrum Disorder/genetics , MicroRNAs/genetics , Olfactory Mucosa/pathology , 3' Untranslated Regions/genetics , Adult , Animals , Astrocytes/metabolism , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/physiopathology , Cells, Cultured , Female , Fibroblasts/metabolism , Genetic Vectors/genetics , Hippocampus/cytology , Hippocampus/embryology , Humans , Lentivirus/genetics , Male , Mice , MicroRNAs/physiology , Neurons/metabolism , Neurons/ultrastructure , Organ Specificity , Real-Time Polymerase Chain Reaction , Transcriptome , Young Adult
18.
Hum Mol Genet ; 24(23): 6736-55, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26376863

ABSTRACT

ATP6AP2, an essential accessory component of the vacuolar H+ ATPase (V-ATPase), has been associated with intellectual disability (ID) and Parkinsonism. ATP6AP2 has been implicated in several signalling pathways; however, little is known regarding its role in the nervous system. To decipher its function in behaviour and cognition, we generated and characterized conditional knockdowns of ATP6AP2 in the nervous system of Drosophila and mouse models. In Drosophila, ATP6AP2 knockdown induced defective phototaxis and vacuolated photoreceptor neurons and pigment cells when depleted in eyes and altered short- and long-term memory when depleted in the mushroom body. In mouse, conditional Atp6ap2 deletion in glutamatergic neurons (Atp6ap2(Camk2aCre/0) mice) caused increased spontaneous locomotor activity and altered fear memory. Both Drosophila ATP6AP2 knockdown and Atp6ap2(Camk2aCre/0) mice presented with presynaptic transmission defects, and with an abnormal number and morphology of synapses. In addition, Atp6ap2(Camk2aCre/0) mice showed autophagy defects that led to axonal and neuronal degeneration in the cortex and hippocampus. Surprisingly, axon myelination was affected in our mutant mice, and axonal transport alterations were observed in Drosophila. In accordance with the identified phenotypes across species, genome-wide transcriptome profiling of Atp6ap2(Camk2aCre/0) mouse hippocampi revealed dysregulation of genes involved in myelination, action potential, membrane-bound vesicles and motor behaviour. In summary, ATP6AP2 disruption in mouse and fly leads to cognitive impairment and neurodegeneration, mimicking aspects of the neuropathology associated with ATP6AP2 mutations in humans. Our results identify ATP6AP2 as an essential gene for the nervous system.


Subject(s)
Cognition Disorders/etiology , Drosophila Proteins/genetics , Membrane Proteins/genetics , Nerve Degeneration/etiology , Proton-Translocating ATPases/genetics , Receptors, Cell Surface/genetics , Animals , Brain/metabolism , Brain/physiopathology , Cognition Disorders/genetics , Cognition Disorders/physiopathology , Disease Models, Animal , Drosophila , Female , Gene Knockdown Techniques , Intellectual Disability/genetics , Male , Mice , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/physiology , Neurons/ultrastructure , Parkinsonian Disorders/genetics , Synapses/metabolism , Synapses/physiology , Synapses/ultrastructure
19.
Brain Struct Funct ; 220(6): 3673-82, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25158900

ABSTRACT

The process of learning mainly depends on the ability to store new information, while the ability to retrieve this information and express appropriate behaviors are also crucial for the adaptation of individuals to environmental cues. Thereby, all three components contribute to the cognitive fitness of an individual. While a lack of behavioral adaptation is a recurrent trait of intellectually disabled patients, discriminating between memory formation, memory retrieval or behavioral expression deficits is not easy to establish. Here, we report some deficits in contextual fear behavior in knockout mice for the intellectual disability gene Il1rapl1. Functional in vivo experiments revealed that the lack of conditioned response resulted from a local inhibitory to excitatory (I/E) imbalance in basolateral amygdala (BLA) consecutive to a loss of excitatory drive onto BLA principal cells by caudal hippocampus axonal projections. A normalization of the fear behavior was obtained in adult mutant mice following opsin-based in vivo synaptic priming of hippocampo-BLA synapses in adult il1rapl1 knockout mice, indicating that synaptic efficacy at hippocampo-BLA projections is crucial for contextual fear memory expression. Importantly, because this restoration was obtained after the learning phase, our results suggest that some of the genetically encoded cognitive deficits in humans may originate from a lack of restitution of genuinely formed memories rather than an exclusive inability to store new memories.


Subject(s)
Amygdala/physiology , Fear/physiology , Hippocampus/physiology , Intellectual Disability/physiopathology , Interleukin-1 Receptor Accessory Protein/physiology , Animals , Conditioning, Classical/physiology , Disease Models, Animal , Intellectual Disability/genetics , Interleukin-1 Receptor Accessory Protein/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Synapses/physiology , Synaptic Potentials
20.
Hum Mol Genet ; 24(4): 1106-18, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25305082

ABSTRACT

Mutations in interleukin-1 receptor accessory protein like 1 (IL1RAPL1) gene have been associated with non-syndromic intellectual disability (ID) and autism spectrum disorder. This protein interacts with synaptic partners like PSD-95 and PTPδ, regulating the formation and function of excitatory synapses. The aim of this work was to characterize the synaptic consequences of three IL1RAPL1 mutations, two novel causing the deletion of exon 6 (Δex6) and one point mutation (C31R), identified in patients with ID. Using immunofluorescence and electrophysiological recordings, we examined the effects of IL1RAPL1 mutant over-expression on synapse formation and function in cultured rodent hippocampal neurons. Δex6 but not C31R mutation leads to IL1RAPL1 protein instability and mislocalization within dendrites. Analysis of different markers of excitatory synapses and sEPSC recording revealed that both mutants fail to induce pre- and post-synaptic differentiation, contrary to WT IL1RAPL1 protein. Cell aggregation and immunoprecipitation assays in HEK293 cells showed a reduction of the interaction between IL1RAPL1 mutants and PTPδ that could explain the observed synaptogenic defect in neurons. However, these mutants do not affect all cellular signaling because their over-expression still activates JNK pathway. We conclude that both mutations described in this study lead to a partial loss of function of the IL1RAPL1 protein through different mechanisms. Our work highlights the important function of the trans-synaptic PTPδ/IL1RAPL1 interaction in synaptogenesis and as such in ID in the patients.


Subject(s)
Intellectual Disability/genetics , Interleukin-1 Receptor Accessory Protein/genetics , Mutation , Neurogenesis/genetics , Synapses/genetics , Adult , Child , Child, Preschool , DNA Mutational Analysis , Exons , Female , Humans , Intellectual Disability/metabolism , Interleukin-1 Receptor Accessory Protein/chemistry , Interleukin-1 Receptor Accessory Protein/metabolism , Introns , Male , Pedigree , Polymorphism, Single Nucleotide , Protein Interaction Domains and Motifs , Protein Transport , Sequence Deletion , Signal Transduction , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL