Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Neurosci ; 43(50): 8744-8755, 2023 12 13.
Article in English | MEDLINE | ID: mdl-37857485

ABSTRACT

Mammalian target of rapamycin (mTOR) pathway has emerged as a key molecular mechanism underlying memory processes. Although mTOR inhibition is known to block memory processes, it remains elusive whether and how an enhancement of mTOR signaling may improve memory processes. Here we found in male mice that the administration of VO-OHpic, an inhibitor of the phosphatase and tensin homolog (PTEN) that negatively modulates AKT-mTOR pathway, enhanced auditory fear memory for days and weeks, while it left short-term memory unchanged. Memory enhancement was associated with a long-lasting increase in immature-type dendritic spines of pyramidal neurons into the auditory cortex. The persistence of spine remodeling over time arose by the interplay between PTEN inhibition and memory processes, as VO-OHpic induced only a transient immature spine growth in the somatosensory cortex, a region not involved in long-term auditory memory. Both the potentiation of fear memories and increase in immature spines were hampered by rapamycin, a selective inhibitor of mTORC1. These data revealed that memory can be potentiated over time by the administration of a selective PTEN inhibitor. In addition to disclosing new information on the cellular mechanisms underlying long-term memory maintenance, our study provides new insights on the molecular processes that aid enhancing memories over time.SIGNIFICANCE STATEMENT The neuronal mechanisms that may help improve the maintenance of long-term memories are still elusive. The inhibition of mammalian-target of rapamycin (mTOR) signaling shows that this pathway plays a crucial role in synaptic plasticity and memory formation. However, whether its activation may strengthen long-term memory storage is unclear. We assessed the consequences of positive modulation of AKT-mTOR pathway obtained by VO-OHpic administration, a phosphatase and tensin homolog inhibitor, on memory retention and underlying synaptic modifications. We found that mTOR activation greatly enhanced memory maintenance for weeks by producing a long-lasting increase of immature-type dendritic spines in pyramidal neurons of the auditory cortex. These results offer new insights on the cellular and molecular mechanisms that can aid enhancing memories over time.


Subject(s)
Auditory Cortex , Proto-Oncogene Proteins c-akt , Male , Mice , Animals , Mechanistic Target of Rapamycin Complex 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Auditory Cortex/metabolism , Dendritic Spines/metabolism , Tensins/metabolism , Memory, Long-Term/physiology , TOR Serine-Threonine Kinases/metabolism , Memory, Short-Term/physiology , Sirolimus/pharmacology , Fear/physiology , Phosphoric Monoester Hydrolases/metabolism , Mammals
2.
Int J Mol Sci ; 23(14)2022 Jul 11.
Article in English | MEDLINE | ID: mdl-35886995

ABSTRACT

Collagen VI-related disorders (COL6-RD) represent a severe form of congenital disease for which there is no treatment. Dominant-negative pathogenic variants in the genes encoding α chains of collagen VI are the main cause of COL6-RD. Here we report that patient-derived fibroblasts carrying a common single nucleotide variant mutation are unable to build the extracellular collagen VI network. This correlates with the intracellular accumulation of endosomes and lysosomes triggered by the increased phosphorylation of the collagen VI receptor CMG2. Notably, using a CRISPR-Cas9 gene-editing tool to silence the dominant-negative mutation in patients' cells, we rescued the normal extracellular collagen VI network, CMG2 phosphorylation levels, and the accumulation of endosomes and lysosomes. Our findings reveal an unanticipated role of CMG2 in regulating endosomal and lysosomal homeostasis and suggest that mutated collagen VI dysregulates the intracellular environment in fibroblasts in collagen VI-related muscular dystrophy.


Subject(s)
Collagen Type VI , Muscular Dystrophies , Receptors, Peptide , Collagen Type VI/genetics , Extracellular Matrix/pathology , Humans , Morphogenesis , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Mutation , Receptors, Peptide/genetics
3.
Antioxidants (Basel) ; 11(1)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35052661

ABSTRACT

One of the most important characteristics of the brain compared to other organs is its elevated metabolic demand. Consequently, neurons consume high quantities of oxygen, generating significant amounts of reactive oxygen species (ROS) as a by-product. These potentially toxic molecules cause oxidative stress (OS) and are associated with many disorders of the nervous system, where pathological processes such as aberrant protein oxidation can ultimately lead to cellular dysfunction and death. Epilepsy, characterized by a long-term predisposition to epileptic seizures, is one of the most common of the neurological disorders associated with OS. Evidence shows that increased neuronal excitability-the hallmark of epilepsy-is accompanied by neuroinflammation and an excessive production of ROS; together, these factors are likely key features of seizure initiation and propagation. This review discusses the role of OS in epilepsy, its connection to neuroinflammation and the impact on synaptic function. Considering that the pharmacological treatment options for epilepsy are limited by the heterogeneity of these disorders, we also introduce the latest advances in anti-epileptic drugs (AEDs) and how they interact with OS. We conclude that OS is intertwined with numerous physiological and molecular mechanisms in epilepsy, although a causal relationship is yet to be established.

4.
Cell Mol Life Sci ; 78(7): 3503-3524, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33340069

ABSTRACT

Members of the Tre2/Bub2/Cdc16 (TBC), lysin motif (LysM), domain catalytic (TLDc) protein family are associated with multiple neurodevelopmental disorders, although their exact roles in disease remain unclear. For example, nuclear receptor coactivator 7 (NCOA7) has been associated with autism, although almost nothing is known regarding the mode-of-action of this TLDc protein in the nervous system. Here we investigated the molecular function of NCOA7 in neurons and generated a novel mouse model to determine the consequences of deleting this locus in vivo. We show that NCOA7 interacts with the cytoplasmic domain of the vacuolar (V)-ATPase in the brain and demonstrate that this protein is required for normal assembly and activity of this critical proton pump. Neurons lacking Ncoa7 exhibit altered development alongside defective lysosomal formation and function; accordingly, Ncoa7 deletion animals exhibited abnormal neuronal patterning defects and a reduced expression of lysosomal markers. Furthermore, behavioural assessment revealed anxiety and social defects in mice lacking Ncoa7. In summary, we demonstrate that NCOA7 is an important V-ATPase regulatory protein in the brain, modulating lysosomal function, neuronal connectivity and behaviour; thus our study reveals a molecular mechanism controlling endolysosomal homeostasis that is essential for neurodevelopment.


Subject(s)
Behavior, Animal , Disease Models, Animal , Neurodevelopmental Disorders/pathology , Neurons/pathology , Nuclear Receptor Coactivators/physiology , Oxidative Stress , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Endosomes/metabolism , Female , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/metabolism , Neurons/metabolism , Vacuolar Proton-Translocating ATPases/genetics
5.
Neurobiol Dis ; 139: 104822, 2020 06.
Article in English | MEDLINE | ID: mdl-32113911

ABSTRACT

DEP-domain containing 5 (DEPDC5) is part of the GATOR1 complex that functions as key inhibitor of the mechanistic target of rapamycin complex 1 (mTORC1). Loss-of-function mutations in DEPDC5 leading to mTOR hyperactivation have been identified as the most common cause of either lesional or non-lesional focal epilepsy. However, the precise mechanisms by which DEPDC5 loss-of-function triggers neuronal and network hyperexcitability are still unclear. In this study, we investigated the cellular mechanisms of hyperexcitability by comparing the constitutive heterozygous Depdc5 knockout mouse versus different levels of acute Depdc5 deletion (≈40% and ≈80% neuronal knockdown of Depdc5 protein) by RNA interference in primary cortical cultures. While heterozygous Depdc5+/- neurons have only a subtle phenotype, acutely knocked-down neurons exhibit a strong dose-dependent phenotype characterized by mTOR hyperactivation, increased soma size, dendritic arborization, excitatory synaptic transmission and intrinsic excitability. The robust synaptic phenotype resulting from the acute knockdown Depdc5 deficiency highlights the importance of the temporal dynamics of Depdc5 knockdown in triggering the phenotypic changes, reminiscent of the somatic second-hit mechanism in patients with focal cortical dysplasia. These findings uncover a novel synaptic phenotype that is causally linked to Depdc5 knockdown, highlighting the developmental role of Depdc5. Interestingly, the synaptic defect appears to affect only excitatory synapses, while inhibitory synapses develop normally. The increased frequency and amplitude of mEPSCs, paralleled by increased density of excitatory synapses and expression of glutamate receptors, may generate an excitation/inhibition imbalance that triggers epileptogenesis.


Subject(s)
Epilepsies, Partial/genetics , GTPase-Activating Proteins/genetics , TOR Serine-Threonine Kinases/genetics , Animals , Disease Models, Animal , Female , Male , Malformations of Cortical Development/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Knockout , Mutation , Phenotype , Repressor Proteins/genetics , Signal Transduction
6.
J Orofac Orthop ; 80(2): 79-87, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30673806

ABSTRACT

PURPOSE: The aims of the study were (1) to evaluate the fitting of three different aligners (Invisalign [Align Technology, Santa Clara, CA, USA], CA Clear Aligner [Scheu-Dental, Iserlohn, Germany] and F22 [Sweden&Martina, Due Carrare, Italy]) on anchorage attachments using scanning electron microscopy (SEM), and (2) to analyze the influence of 2 different types of resin used to build attachments on aligner fitting. METHODS: Using STL files of a patient, six resin casts were obtained and rectangular attachments were bonded on them. Conventional bulk-fill resin was used to build upper attachments while a flowable resin was used to build the lower ones. Passive aligners were adapted on each cast and then sectioned buccolingually. Microphotographs of the obtained sections were performed using a SEM and then micrometric measurements of aligner fitting on anchorage attachments were recorded. RESULTS: Analyzing the overall fitting of upper arch aligners, Invisalign provided a significantly better fitting with respect to F22 (P = 0.009); differences were not significant when comparing Invisalign with CA Clear Aligner, and CA Clear Aligner with F22. Analyzing the overall fitting of lower arch aligners, F22 provided a significantly better fitting with respect to CA Clear Aligner (P = 0.008) and Invisalign (P = 0.011). The analysis showed a significantly better fitting on upper attachments, built using conventional bulk-fill resin (P = 0.034). CONCLUSIONS: Invisalign, CA Clear Aligner and F22 have comparable performance in terms of fitting on anchorage attachments. Conventional bulk-fill resin provides the best fitting on anchorage attachments.


Subject(s)
Orthodontic Anchorage Procedures , Orthodontic Appliances, Removable , Dental Casting Technique , Humans , Malocclusion, Angle Class I/therapy , Microscopy, Electron, Scanning , Resins, Synthetic/therapeutic use , Tooth Movement Techniques/instrumentation , Tooth Movement Techniques/methods
7.
Dis Model Mech ; 12(2)2019 02 22.
Article in English | MEDLINE | ID: mdl-30692144

ABSTRACT

Loss-of-function mutations in a human AMPA receptor-associated protein, ferric chelate reductase 1-like (FRRS1L), are associated with a devastating neurological condition incorporating choreoathetosis, cognitive deficits and epileptic encephalopathies. Furthermore, evidence from overexpression and ex vivo studies has implicated FRRS1L in AMPA receptor biogenesis, suggesting that changes in glutamatergic signalling might underlie the disorder. Here, we investigated the neurological and neurobehavioural correlates of the disorder using a mouse Frrs1l null mutant. The study revealed several neurological defects that mirrored those seen in human patients. We established that mice lacking Frrs1l suffered from a broad spectrum of early-onset motor deficits with no progressive, age-related deterioration. Moreover, Frrs1l-/- mice were hyperactive, irrespective of test environment, exhibited working memory deficits and displayed significant sleep fragmentation. Longitudinal electroencephalographic (EEG) recordings also revealed abnormal EEG results in Frrs1l-/- mice. Parallel investigations into disease aetiology identified a specific deficiency in AMPA receptor levels in the brain of Frrs1l-/- mice, while the general levels of several other synaptic components remained unchanged, with no obvious alterations in the number of synapses. Furthermore, we established that Frrsl1 deletion results in an increased proportion of immature AMPA receptors, indicated by incomplete glycosylation of GLUA2 (also known as GRIA2) and GLUA4 (also known as GRIA4) AMPA receptor proteins. This incomplete maturation leads to cytoplasmic retention and a reduction of those specific AMPA receptor levels in the postsynaptic membrane. Overall, this study determines, for the first time in vivo, how loss of FRRS1L function can affect glutamatergic signalling, and provides mechanistic insight into the development and progression of a human hyperkinetic disorder.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cognition , Electrophysiological Phenomena , Membrane Proteins/metabolism , Motor Activity , Nerve Tissue Proteins/metabolism , Nervous System/growth & development , Nervous System/pathology , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Animals, Newborn , Body Size , Brain/metabolism , Brain/pathology , Cognition Disorders/pathology , Cytoplasm/metabolism , Glycosylation , Membrane Proteins/genetics , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nervous System/physiopathology , Sleep , Survival Analysis
8.
Hum Mol Genet ; 28(4): 584-597, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30335140

ABSTRACT

Mutations in the Tre2/Bub2/Cdc16 (TBC)1 domain family member 24 (TBC1D24) gene are associated with a range of inherited neurological disorders, from drug-refractory lethal epileptic encephalopathy and DOORS syndrome (deafness, onychodystrophy, osteodystrophy, mental retardation, seizures) to non-syndromic hearing loss. TBC1D24 has been implicated in neuronal transmission and maturation, although the molecular function of the gene and the cause of the apparently complex disease spectrum remain unclear. Importantly, heterozygous TBC1D24 mutation carriers have also been reported with seizures, suggesting that haploinsufficiency for TBC1D24 is significant clinically. Here we have systematically investigated an allelic series of disease-associated mutations in neurons alongside a new mouse model to investigate the consequences of TBC1D24 haploinsufficiency to mammalian neurodevelopment and synaptic physiology. The cellular studies reveal that disease-causing mutations that disrupt either of the conserved protein domains in TBC1D24 are implicated in neuronal development and survival and are likely acting as loss-of-function alleles. We then further investigated TBC1D24 haploinsufficiency in vivo and demonstrate that TBC1D24 is also crucial for normal presynaptic function: genetic disruption of Tbc1d24 expression in the mouse leads to an impairment of endocytosis and an enlarged endosomal compartment in neurons with a decrease in spontaneous neurotransmission. These data reveal the essential role for TBC1D24 at the mammalian synapse and help to define common synaptic mechanisms that could underlie the varied effects of TBC1D24 mutations in neurological disease.


Subject(s)
Carrier Proteins/genetics , Craniofacial Abnormalities/genetics , Epilepsy/genetics , Hand Deformities, Congenital/genetics , Hearing Loss, Sensorineural/genetics , Intellectual Disability/genetics , Nails, Malformed/genetics , Seizures/genetics , Amino Acid Sequence/genetics , Animals , Craniofacial Abnormalities/physiopathology , Disease Models, Animal , Endocytosis/genetics , Epilepsy/physiopathology , Exome/genetics , GTPase-Activating Proteins , Gene Expression Regulation , Hand Deformities, Congenital/physiopathology , Haploinsufficiency , Hearing Loss, Sensorineural/physiopathology , Humans , Intellectual Disability/physiopathology , Membrane Proteins , Mice , Mutation , Nails, Malformed/physiopathology , Nerve Tissue Proteins , Neuronal Plasticity/genetics , Neurons/metabolism , Neurons/pathology , Pedigree , Seizures/physiopathology
9.
Cereb Cortex ; 29(5): 2010-2033, 2019 05 01.
Article in English | MEDLINE | ID: mdl-29912316

ABSTRACT

Mutations in PRoline-Rich Transmembrane protein 2 (PRRT2) underlie a group of paroxysmal disorders including epilepsy, kinesigenic dyskinesia and migraine. Most of the mutations lead to impaired PRRT2 expression and/or function, emphasizing the pathogenic role of the PRRT2 deficiency. In this work, we investigated the phenotype of primary hippocampal neurons obtained from mouse embryos in which the PRRT2 gene was constitutively inactivated. Although PRRT2 is expressed by both excitatory and inhibitory neurons, its deletion decreases the number of excitatory synapses without significantly affecting the number of inhibitory synapses or the nerve terminal ultrastructure. Analysis of synaptic function in primary PRRT2 knockout excitatory neurons by live imaging and electrophysiology showed slowdown of the kinetics of exocytosis, weakened spontaneous and evoked synaptic transmission and markedly increased facilitation. Inhibitory neurons showed strengthening of basal synaptic transmission, accompanied by faster depression. At the network level these complex synaptic effects resulted in a state of heightened spontaneous and evoked activity that was associated with increased excitability of excitatory neurons in both PRRT2 knockout primary cultures and acute hippocampal slices. The data indicate the existence of network instability/hyperexcitability as the possible basis of the paroxysmal phenotypes associated with PRRT2 mutations.


Subject(s)
Hippocampus/physiology , Membrane Proteins/physiology , Neuronal Plasticity , Neurons/physiology , Synaptic Transmission , Animals , Cells, Cultured , Exocytosis , Male , Membrane Potentials , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/physiology , Synapses/physiology , Synapses/ultrastructure
10.
Angle Orthod ; 88(5): 596-601, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29911907

ABSTRACT

OBJECTIVES: The fitting of aligners on anchorage teeth is a crucial factor in clear aligner orthodontics. The purpose of this experimental study was to evaluate the fitting of two aligner systems, Invisalign and CA-Clear Aligner, using scanning electron microscopy (SEM). MATERIALS AND METHODS: Passive aligners (Invisalign and CA-Clear Aligner) were adapted on resin casts obtained by stereolithography (STL) files of a patient, and then sectioned buccolingually. Upper and lower central incisors, upper and lower first premolars, and upper and lower first molars were the regions analyzed. Representative microphotographs of sections were taken with a scanning electron microscope (SEM); a total of 160 micrometric measurements were obtained and analyzed with ANOVA tests. RESULTS: Invisalign provided an overall better fit on lower incisors ( F = 11.48, P = .0095) and on lower molars ( F = 19.93, P = .0012). Considering the different regions, Invisalign provided better fit at the gingival edge of the buccal aspect on lower incisors ( F = 11.33, P = 0.0056) and at the gingival edge of the lingual aspect on upper premolars ( F =5.34, P = 0.0047). On the upper molars, Invisalign provided better fit at the gingival edge of the buccal aspect, while CA-Clear Aligner provided better fit at the buccal maximum convexity, on the buccal cusp, on the occlusal groove and at the palatal cusp. On lower molars, Invisalign showed a more accurate fit at the buccal aspect points. CONCLUSIONS: Invisalign and CA-Clear Aligner exhibited comparable fit on anchorage teeth. Invisalign provided better fit at the gingival edges of aligners, while the CA-Clear Aligner provided better fit on complex occlusal surfaces.


Subject(s)
Orthodontic Appliances, Removable , Dental Casting Technique , Humans , Microscopy, Electron, Scanning , Stereolithography
12.
Sci Rep ; 7(1): 16825, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203796

ABSTRACT

Down syndrome (DS) is caused by the triplication of human chromosome 21 and represents the most frequent genetic cause of intellectual disability. The trisomic Ts65Dn mouse model of DS shows synaptic deficits and reproduces the essential cognitive disabilities of the human syndrome. Aerobic exercise improved various neurophysiological dysfunctions in Ts65Dn mice, including hippocampal synaptic deficits, by promoting synaptogenesis and neurotransmission at glutamatergic terminals. Most importantly, the same intervention also prompted the recovery of hippocampal adult neurogenesis and synaptic plasticity and restored cognitive performance in trisomic mice. Additionally, the expression of brain-derived neurotrophic factor (BDNF) was markedly decreased in the hippocampus of patients with DS. Since the positive effect of exercise was paralleled by increased BDNF expression in trisomic mice, we investigated the effectiveness of a BDNF-mimetic treatment with 7,8-dihydroxyflavone at alleviating intellectual disabilities in the DS model. Pharmacological stimulation of BDNF signaling rescued synaptic plasticity and memory deficits in Ts65Dn mice. Based on our findings, Ts65Dn mice benefit from interventions aimed at promoting brain plasticity, and we provide evidence that BDNF signaling represents a potentially new pharmacological target for treatments aimed at rescuing cognitive disabilities in patients with DS.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Down Syndrome/pathology , Flavones/pharmacology , Learning/drug effects , Memory/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Down Syndrome/drug therapy , Excitatory Postsynaptic Potentials/drug effects , Female , Flavones/therapeutic use , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis , Neuronal Plasticity/drug effects , Physical Conditioning, Animal , Signal Transduction/drug effects
13.
Cell Rep ; 21(12): 3596-3611, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29262337

ABSTRACT

Synaptic transmission is critically dependent on synaptic vesicle (SV) recycling. Although the precise mechanisms of SV retrieval are still debated, it is widely accepted that a fundamental role is played by clathrin-mediated endocytosis, a form of endocytosis that capitalizes on the clathrin/adaptor protein complex 2 (AP2) coat and several accessory factors. Here, we show that the previously uncharacterized protein KIAA1107, predicted by bioinformatics analysis to be involved in the SV cycle, is an AP2-interacting clathrin-endocytosis protein (APache). We found that APache is highly enriched in the CNS and is associated with clathrin-coated vesicles via interaction with AP2. APache-silenced neurons exhibit a severe impairment of maturation at early developmental stages, reduced SV density, enlarged endosome-like structures, and defects in synaptic transmission, consistent with an impaired clathrin/AP2-mediated SV recycling. Our data implicate APache as an actor in the complex regulation of SV trafficking, neuronal development, and synaptic plasticity.


Subject(s)
Adaptor Protein Complex 2 , Endocytosis , Neurogenesis , Synaptic Vesicles/metabolism , Adaptor Protein Complex 2/metabolism , Animals , Cells, Cultured , Clathrin-Coated Vesicles/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Protein Binding , Rats , Rats, Sprague-Dawley
14.
Neurobiol Dis ; 99: 66-83, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28007585

ABSTRACT

Heterozygous and rare homozygous mutations in PRoline-Rich Transmembrane protein 2 (PRRT2) underlie a group of paroxysmal disorders including epilepsy, kinesigenic dyskinesia episodic ataxia and migraine. Most of the mutations lead to impaired PRRT2 expression and/or function. Recently, an important role for PRTT2 in the neurotransmitter release machinery, brain development and synapse formation has been uncovered. In this work, we have characterized the phenotype of a mouse in which the PRRT2 gene has been constitutively inactivated (PRRT2 KO). ß-galactosidase staining allowed to map the regional expression of PRRT2 that was more intense in the cerebellum, hindbrain and spinal cord, while it was localized to restricted areas in the forebrain. PRRT2 KO mice are normal at birth, but display paroxysmal movements at the onset of locomotion that persist in the adulthood. In addition, adult PRRT2 KO mice present abnormal motor behaviors characterized by wild running and jumping in response to audiogenic stimuli that are ineffective in wild type mice and an increased sensitivity to the convulsive effects of pentylentetrazol. Patch-clamp electrophysiology in hippocampal and cerebellar slices revealed specific effects in the cerebellum, where PRRT2 is highly expressed, consisting in a higher excitatory strength at parallel fiber-Purkinje cell synapses during high frequency stimulation. The results show that the PRRT2 KO mouse reproduces the motor paroxysms present in the human PRRT2-linked pathology and can be proposed as an experimental model for the study of the pathogenesis of the disease as well as for testing personalized therapeutic approaches.


Subject(s)
Brain/physiopathology , Membrane Proteins/deficiency , Motor Activity/physiology , Motor Disorders/physiopathology , Seizures/physiopathology , Animals , Animals, Newborn , Brain/growth & development , Brain/pathology , Cognition/physiology , Disease Models, Animal , Female , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Motor Disorders/pathology , Mutation , Nerve Tissue Proteins/genetics , Pentylenetetrazole , Phenotype , Seizures/pathology , Spinal Cord/growth & development , Spinal Cord/pathology , Spinal Cord/physiopathology , Synapses/pathology , Synapses/physiology , Tissue Culture Techniques
15.
Front Cell Neurosci ; 10: 261, 2016.
Article in English | MEDLINE | ID: mdl-27965538

ABSTRACT

Cyclin-dependent kinase-like 5 (CDKL5) mutations are found in severe neurodevelopmental disorders, including the Hanefeld variant of Rett syndrome (RTT; CDKL5 disorder). CDKL5 loss-of-function murine models recapitulate pathological signs of the human disease, such as visual attention deficits and reduced visual acuity. Here we investigated the cellular and synaptic substrates of visual defects by studying the organization of the primary visual cortex (V1) of Cdkl5-/y mice. We found a severe reduction of c-Fos expression in V1 of Cdkl5-/y mutants, suggesting circuit hypoactivity. Glutamatergic presynaptic structures were increased, but postsynaptic PSD-95 and Homer were significantly downregulated in CDKL5 mutants. Interneurons expressing parvalbumin, but not other types of interneuron, had a higher density in mutant V1, and were hyperconnected with pyramidal neurons. Finally, the developmental trajectory of pavalbumin-containing cells was also affected in Cdkl5-/y mice, as revealed by fainter appearance perineuronal nets at the closure of the critical period (CP). The present data reveal an overall disruption of V1 cellular and synaptic organization that may cause a shift in the excitation/inhibition balance likely to underlie the visual deficits characteristic of CDKL5 disorder. Moreover, ablation of CDKL5 is likely to tamper with the mechanisms underlying experience-dependent refinement of cortical circuits during the CP of development.

16.
Cell Rep ; 15(1): 117-131, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27052163

ABSTRACT

Heterozygous mutations in proline-rich transmembrane protein 2 (PRRT2) underlie a group of paroxysmal disorders, including epilepsy, kinesigenic dyskinesia, and migraine. Most of the mutations lead to impaired PRRT2 expression, suggesting that loss of PRRT2 function may contribute to pathogenesis. We show that PRRT2 is enriched in presynaptic terminals and that its silencing decreases the number of synapses and increases the number of docked synaptic vesicles at rest. PRRT2-silenced neurons exhibit a severe impairment of synchronous release, attributable to a sharp decrease in release probability and Ca(2+) sensitivity and associated with a marked increase of the asynchronous/synchronous release ratio. PRRT2 interacts with the synaptic proteins SNAP-25 and synaptotagmin 1/2. The results indicate that PRRT2 is intimately connected with the Ca(2+)-sensing machinery and that it plays an important role in the final steps of neurotransmitter release.


Subject(s)
Calcium Signaling , Exocytosis , Membrane Proteins/metabolism , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Animals , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Presynaptic Terminals/physiology , Rats , Rats, Sprague-Dawley , Synaptic Potentials , Synaptic Vesicles/metabolism , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmins/metabolism
17.
J Biol Chem ; 291(12): 6111-23, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26797119

ABSTRACT

Proline-rich transmembrane protein 2 (PRRT2) has been identified as the single causative gene for a group of paroxysmal syndromes of infancy, including epilepsy, paroxysmal movement disorders, and migraine. On the basis of topology predictions, PRRT2 has been assigned to the recently characterized family of Dispanins, whose members share the two-transmembrane domain topology with a large N terminus and short C terminus oriented toward the outside of the cell. Because PRRT2 plays a role at the synapse, it is important to confirm the exact orientation of its N and C termini with respect to the plasma membrane to get clues regarding its possible function. Using a combination of different experimental approaches, including live immunolabeling, immunogold electron microscopy, surface biotinylation and computational modeling, we demonstrate a novel topology for this protein. PRRT2 is a type II transmembrane protein in which only the second hydrophobic segment spans the plasma membrane, whereas the first one is associated with the internal surface of the membrane and forms a helix-loop-helix structure without crossing it. Most importantly, the large proline-rich N-terminal domain is not exposed to the extracellular space but is localized intracellularly, and only the short C terminus is extracellular (N cyt/C exo topology). Accordingly, we show that PRRT2 interacts with the Src homology 3 domain-bearing protein Intersectin 1, an intracellular protein involved in synaptic vesicle cycling. These findings will contribute to the clarification of the role of PRRT2 at the synapse and the understanding of pathogenic mechanisms on the basis of PRRT2-related neurological disorders.


Subject(s)
Membrane Proteins/metabolism , Synapses/metabolism , Animals , Biotinylation , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Membrane Proteins/chemistry , Mice , Molecular Dynamics Simulation , Protein Processing, Post-Translational , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Synaptosomes/metabolism
18.
PLoS One ; 9(5): e91613, 2014.
Article in English | MEDLINE | ID: mdl-24838000

ABSTRACT

Mutations in cyclin-dependent kinase-like 5 (CDKL5) cause early-onset epileptic encephalopathy, a neurodevelopmental disorder with similarities to Rett Syndrome. Here we describe the physiological, molecular, and behavioral phenotyping of a Cdkl5 conditional knockout mouse model of CDKL5 disorder. Behavioral analysis of constitutive Cdkl5 knockout mice revealed key features of the human disorder, including limb clasping, hypoactivity, and abnormal eye tracking. Anatomical, physiological, and molecular analysis of the knockout uncovered potential pathological substrates of the disorder, including reduced dendritic arborization of cortical neurons, abnormal electroencephalograph (EEG) responses to convulsant treatment, decreased visual evoked responses (VEPs), and alterations in the Akt/rpS6 signaling pathway. Selective knockout of Cdkl5 in excitatory and inhibitory forebrain neurons allowed us to map the behavioral features of the disorder to separable cell-types. These findings identify physiological and molecular deficits in specific forebrain neuron populations as possible pathological substrates in CDKL5 disorder.


Subject(s)
Disease Models, Animal , Phenotype , Prosencephalon/pathology , Protein Serine-Threonine Kinases/genetics , Rett Syndrome/genetics , Rett Syndrome/pathology , Signal Transduction/physiology , Spasms, Infantile/genetics , Spasms, Infantile/pathology , Analysis of Variance , Animals , Blotting, Western , Dendrites/pathology , Electroencephalography , Epileptic Syndromes , Evoked Potentials, Visual/physiology , Eye Movements/physiology , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...