Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 22337, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38102151

ABSTRACT

We study the role of contingent convertible bonds (CoCos) in a complex network of interconnected banks. By studying the system's phase transitions, we reveal that the structure of the interbank network is of fundamental importance for the effectiveness of CoCos as a financial stability enhancing mechanism. Our results show that, under some network structures, the presence of CoCos can increase (and not reduce) financial fragility, because of the occurring of unneeded triggers and consequential suboptimal conversions that damage CoCos investors. We also demonstrate that, in the presence of a moderate financial shock, lightly interconnected financial networks are more robust than highly interconnected networks. This makes them a potentially optimal choice for both CoCos issuers and buyers.

2.
Mol Autism ; 14(1): 28, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37528484

ABSTRACT

BACKGROUND: Phelan-McDermid syndrome (PMS) is a neurodevelopmental disorder characterized by developmental delay, intellectual disability, and autistic-like behaviors and is primarily caused by haploinsufficiency of SHANK3 gene. Currently, there is no specific treatment for PMS, highlighting the need for a better understanding of SHANK3 functions and the underlying pathophysiological mechanisms in the brain. We hypothesize that SHANK3 haploinsufficiency may lead to alterations in the inhibitory system, which could be linked to the excitatory/inhibitory imbalance observed in models of autism spectrum disorder (ASD). Investigation of these neuropathological features may shed light on the pathogenesis of PMS and potential therapeutic interventions. METHODS: We recorded local field potentials and visual evoked responses in the visual cortex of Shank3∆11-/- mice. Then, to understand the impact of Shank3 in inhibitory neurons, we generated Pv-cre+/- Shank3Fl/Wt conditional mice, in which Shank3 was deleted in parvalbumin-positive neurons. We characterized the phenotype of this murine model and we compared this phenotype before and after ganaxolone administration. RESULTS: We found, in the primary visual cortex, an alteration of the gain control of Shank3 KO compared with Wt mice, indicating a deficit of inhibition on pyramidal neurons. This alteration was rescued after the potentiation of GABAA receptor activity by Midazolam. Behavioral analysis showed an impairment in grooming, memory, and motor coordination of Pv-cre+/- Shank3Fl/Wt compared with Pv-cre+/- Shank3Wt/Wt mice. These deficits were rescued with ganaxolone, a positive modulator of GABAA receptors. Furthermore, we demonstrated that treatment with ganaxolone also ameliorated evocative memory deficits and repetitive behavior of Shank3 KO mice. LIMITATIONS: Despite the significant findings of our study, some limitations remain. Firstly, the neurobiological mechanisms underlying the link between Shank3 deletion in PV neurons and behavioral alterations need further investigation. Additionally, the impact of Shank3 on other classes of inhibitory neurons requires further exploration. Finally, the pharmacological activity of ganaxolone needs further characterization to improve our understanding of its potential therapeutic effects. CONCLUSIONS: Our study provides evidence that Shank3 deletion leads to an alteration in inhibitory feedback on cortical pyramidal neurons, resulting in cortical hyperexcitability and ASD-like behavioral problems. Specifically, cell type-specific deletion of Shank3 in PV neurons was associated with these behavioral deficits. Our findings suggest that ganaxolone may be a potential pharmacological approach for treating PMS, as it was able to rescue the behavioral deficits in Shank3 KO mice. Overall, our study highlights the importance of investigating the role of inhibitory neurons and potential therapeutic interventions in neurodevelopmental disorders such as PMS.


Subject(s)
Autism Spectrum Disorder , Problem Behavior , Mice , Animals , Autism Spectrum Disorder/genetics , Nerve Tissue Proteins/genetics , Neurons , Microfilament Proteins
3.
J Pers Med ; 13(3)2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36983719

ABSTRACT

Astrocytes coordinate several homeostatic processes of the central nervous system and play essential roles for normal brain development and response to disease conditions. Protocols for the conversion of human induced pluripotent stem cells (hiPSCs) into mature astrocytes have opened to the generation of in vitro systems to explore astrocytes' functions in living human cell contexts and patient-specific settings. In this study, we present an optimized monolayer procedure to commit hiPSC-derived cortical progenitors into enriched populations of cortical astrocyte progenitor cells (CX APCs) that can be further amplified and efficiently differentiated into mature astrocytes. Our optimized system provides a valid tool to explore the role of these cells in neurodevelopmental and neuropsychiatric diseases, opening it up to applications in drug development and biomarkers discovery/validation.

4.
Eur J Med Genet ; 66(6): 104746, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36967043

ABSTRACT

Phelan-McDermid syndrome (PMS) is a 22q13.3 deletion syndrome that presents with a disturbed development, neurological and psychiatric characteristics, and sometimes other comorbidities like seizures. The epilepsy manifests itself in a variety of seizure semiologies. Further diagnostics using electroencephalogram (EEG) and brain magnetic resonance imaging (MRI) are important in conjunction with the clinical picture of the seizures to decide whether anticonvulsant therapy is necessary. As part of the development of European consensus guidelines we focussed on the prevalence and semiology of epileptic seizures in PMS associated with a pathogenic variant in the SHANK3 gene or the 22q13 deletion involving SHANK3, in order to then be able to make recommendations regarding diagnosis and therapy.


Subject(s)
Chromosome Disorders , Epilepsy , Humans , Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Chromosome Deletion , Epilepsy/diagnosis , Epilepsy/genetics , Seizures/genetics , Chromosomes, Human, Pair 22/genetics
5.
J Pers Med ; 12(9)2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36143309

ABSTRACT

The advent of intra-nasal esketamine (ESK), one of the first so called fast-acting antidepressant, promises to revolutionize the management of treatment resistant depression (TRD). This NMDA receptor antagonist has proven to be rapidly effective in the short- and medium-term course of the illness, revealing its potential in targeting response in TRD. Although many TRD ESK responders are able to achieve remission, a considerable portion of them undergo a metamorphosis of their depression into different clinical presentations, characterized by instable responses and high recurrence rates that can be considered closer to the concept of Difficult to Treat Depression (DTD) than to TRD. The management of these DTD patients usually requires a further complex multidisciplinary approach and can benefit from the valuable contribution of new personalized medicine tools such as therapeutic drug monitoring and pharmacogenetics. Despite this, these patients usually come with long and complex previous treatments history and, often, advanced and sophisticated ongoing pharmacological schemes that can make the finding of new alternative options to face the current recurrences extremely challenging. In this paper, we describe two DTD patients-already receiving intranasal ESK but showing an instable course-who were clinically stabilized by the association with minocycline, a semisynthetic second-generation tetracycline with known and promising antidepressant properties.

6.
Mol Psychiatry ; 27(12): 4994-5006, 2022 12.
Article in English | MEDLINE | ID: mdl-36100669

ABSTRACT

Members of the Shank protein family are master scaffolds of the postsynaptic architecture and mutations within the SHANK genes are causally associated with autism spectrum disorders (ASDs). We generated a Shank2-Shank3 double knockout mouse that is showing severe autism related core symptoms, as well as a broad spectrum of comorbidities. We exploited this animal model to identify cortical brain areas linked to specific autistic traits by locally deleting Shank2 and Shank3 simultaneously. Our screening of 10 cortical subregions revealed that a Shank2/3 deletion within the retrosplenial area severely impairs social memory, a core symptom of ASD. Notably, DREADD-mediated neuronal activation could rescue the social impairment triggered by Shank2/3 depletion. Data indicate that the retrosplenial area has to be added to the list of defined brain regions that contribute to the spectrum of behavioural alterations seen in ASDs.


Subject(s)
Autism Spectrum Disorder , Gyrus Cinguli , Social Interaction , Animals , Mice , Autism Spectrum Disorder/genetics , Microfilament Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Neurons/physiology , Gyrus Cinguli/metabolism , Gyrus Cinguli/pathology
7.
Mol Ther ; 30(7): 2474-2490, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35390543

ABSTRACT

The development of new therapeutic avenues that target the early stages of Alzheimer's disease (AD) is urgently necessary. A disintegrin and metalloproteinase domain 10 (ADAM10) is a sheddase that is involved in dendritic spine shaping and limits the generation of amyloid-ß. ADAM10 endocytosis increases in the hippocampus of AD patients, resulting in the decreased postsynaptic localization of the enzyme. To restore this altered pathway, we developed a cell-permeable peptide (PEP3) with a strong safety profile that is able to interfere with ADAM10 endocytosis, upregulating the postsynaptic localization and activity of ADAM10. After extensive validation, experiments in a relevant animal model clarified the optimal timing of the treatment window. PEP3 administration was effective for the rescue of cognitive defects in APP/PS1 mice only if administered at an early disease stage. Increased ADAM10 activity promoted synaptic plasticity, as revealed by changes in the molecular compositions of synapses and the spine morphology. Even though further studies are required to evaluate efficacy and safety issues of long-term administration of PEP3, these results provide preclinical evidence to support the therapeutic potential of PEP3 in AD.


Subject(s)
Alzheimer Disease , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Endocytosis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Synapses/metabolism
8.
Riv Psichiatr ; 57(2): 57-66, 2022.
Article in Italian | MEDLINE | ID: mdl-35426424

ABSTRACT

About a third of patients with major depressive disorder (MDD) do not have an adequate response to first-line antidepressant treatment, i.e., develop a treatment-resistant depression (TRD). The partial understanding of MDD pathophysiology currently constitutes the major barrier to clinical and research progress on this topic. However, recent advances in genome editing techniques as well as in induced pluripotent stem cells (iPSC) technology are offering unprecedented opportunities in both human disease modelling and drug discovery. These technology progresses have been enabling to set up disease-relevant patient-specific in vitro disease modeling for various mental disorders. The resulting models have the potential to significantly improve pathophysiologic understanding of MDD and then overcome some limitations inherent to animal and post-mortem models. More recently, psychiatry started to deal with the fast acting antidepressant ketamine and its derivates. Although ketamine appears to have the potential to transform the treatment of depression, its specific mechanisms of action are only partially known. Such knowledge is necessary to develop a model to understand the mechanisms behind fast-acting antidepressants, which may enable the discovery of novel glutamatergic compounds for the treatment of MDD. After discussing both the current understanding of ketamine's mechanisms of action, and the state of the art of human iPSC technology, the authors will introduce the implementation of a TRD model based on iPSC human technology and aimed at studying the ketamine's fast acting antidepressant mechanisms of action.


Subject(s)
Depressive Disorder, Major , Depressive Disorder, Treatment-Resistant , Ketamine , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Depressive Disorder, Treatment-Resistant/drug therapy , Humans , Ketamine/pharmacology , Ketamine/therapeutic use
9.
Orphanet J Rare Dis ; 17(1): 27, 2022 01 29.
Article in English | MEDLINE | ID: mdl-35093143

ABSTRACT

Phelan-McDermid syndrome (PMS) was initially called the 22q13 deletion syndrome based on its etiology as a deletion of the distal long arm of chromosome 22. These included terminal and interstitial deletions, as well as other structural rearrangements. Later, pathogenetic variants and deletions of the SHANK3 gene were found to result in a phenotype consistent with PMS. The association between SHANK3 and PMS led investigators to consider disruption/deletion of SHANK3 to be a prerequisite for diagnosing PMS. This narrow definition of PMS based on the involvement of SHANK3 has the adverse effect of causing patients with interstitial deletions of chromosome 22 to "lose" their diagnosis. It also results in underreporting of individuals with interstitial deletions of 22q13 that preserve SHANK3. To reduce the confusion for families, clinicians, researchers, and pharma, a simple classification for PMS has been devised. PMS and will be further classified as PMS-SHANK3 related or PMS-SHANK3 unrelated. PMS can still be used as a general term, but this classification system is inclusive. It allows researchers, regulatory agencies, and other stakeholders to define SHANK3 alterations or interstitial deletions not affecting the SHANK3 coding region.


Subject(s)
Chromosome Disorders , Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 22/genetics , Humans , Phenotype
10.
Mol Autism ; 13(1): 1, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34980259

ABSTRACT

BACKGROUND: Dravet Syndrome is a severe childhood pharmaco-resistant epileptic disorder mainly caused by mutations in the SCN1A gene, which encodes for the α1 subunit of the type I voltage-gated sodium channel (NaV1.1), that causes imbalance between excitation and inhibition in the brain. We recently found that eEF2K knock out mice displayed enhanced GABAergic transmission and tonic inhibition and were less susceptible to epileptic seizures. Thus, we investigated the effect of inhibition of eEF2K on the epileptic and behavioral phenotype of Scn1a ± mice, a murine model of Dravet Syndrome. METHODS: To elucidate the role of eEF2K pathway in the etiopathology of Dravet syndrome we generated a new mouse model deleting the eEF2K gene in Scn1a ± mice. By crossing Scn1a ± mice with eEF2K-/- mice we obtained the three main genotypes needed for our studies, Scn1a+/+ eEF2K+/+ (WT mice), Scn1a ± eEF2K+/+ mice (Scn1a ± mice) and Scn1a ± eEF2K-/- mice, that were fully characterized for EEG and behavioral phenotype. Furthermore, we tested the ability of a pharmacological inhibitor of eEF2K in rescuing EEG alterations of the Scn1a ± mice. RESULTS: We showed that the activity of eEF2K/eEF2 pathway was enhanced in Scn1a ± mice. Then, we demonstrated that both genetic deletion and pharmacological inhibition of eEF2K were sufficient to ameliorate the epileptic phenotype of Scn1a ± mice. Interestingly we also found that motor coordination defect, memory impairments, and stereotyped behavior of the Scn1a ± mice were reverted by eEF2K deletion. The analysis of spontaneous inhibitory postsynaptic currents (sIPSCs) suggested that the rescue of the pathological phenotype was driven by the potentiation of GABAergic synapses. LIMITATIONS: Even if we found that eEF2K deletion was able to increase inhibitory synapses function, the molecular mechanism underlining the inhibition of eEF2K/eEF2 pathway in rescuing epileptic and behavioral alterations in the Scn1a ± needs further investigations. CONCLUSIONS: Our data indicate that pharmacological inhibition of eEF2K could represent a novel therapeutic intervention for treating epilepsy and related comorbidities in the Dravet syndrome.


Subject(s)
Epilepsies, Myoclonic , Epilepsy , Animals , Disease Models, Animal , Elongation Factor 2 Kinase/genetics , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/therapy , Epileptic Syndromes , Mice , Mice, Inbred C57BL , NAV1.1 Voltage-Gated Sodium Channel/genetics
11.
Cell Calcium ; 100: 102480, 2021 12.
Article in English | MEDLINE | ID: mdl-34607180

ABSTRACT

Calcineurin (CaN), acting downstream of intracellular calcium signals, orchestrates cellular remodeling in many cellular types. In astrocytes, major homeostatic players in the central nervous system (CNS), CaN is involved in neuroinflammation and gliosis, while its role in healthy CNS or in early neuro-pathogenesis is poorly understood. Here we report that in mice with conditional deletion of CaN in GFAP-expressing astrocytes (astroglial calcineurin KO, ACN-KO), at 1 month of age, transcription was largely unchanged, while the proteome was deranged in the hippocampus and cerebellum. Gene ontology analysis revealed overrepresentation of annotations related to myelin sheath, mitochondria, ribosome and cytoskeleton. Over-represented pathways were related to protein synthesis, oxidative phosphorylation, mTOR and neurological disorders, including Alzheimer's disease (AD) and seizure disorder. Comparison with published proteomic datasets showed significant overlap with the proteome of a familial AD mouse model and of human subjects with drug-resistant seizures. ACN-KO mice showed no alterations of motor activity, equilibrium, anxiety or depressive state. However, in Barnes maze ACN-KO mice learned the task but adopted serial search strategy. Strikingly, beginning from about 5 months of age ACN-KO mice developed spontaneous tonic-clonic seizures with an inflammatory signature of epileptic brains. Altogether, our data suggest that the deletion of astroglial CaN produces features of neurological disorders and predisposes mice to seizures. We suggest that calcineurin in astrocytes may serve as a novel Ca2+-sensitive switch which regulates protein expression and homeostasis in the central nervous system.


Subject(s)
Alzheimer Disease , Epilepsy , Alzheimer Disease/genetics , Animals , Astrocytes , Calcineurin , Epilepsy/genetics , Mice , Neuroinflammatory Diseases , Proteome , Proteomics , Seizures/genetics
12.
J Neurochem ; 159(1): 12-14, 2021 10.
Article in English | MEDLINE | ID: mdl-34252196

ABSTRACT

Various neuroimaging approaches have reported alterations in brain connectivity in patients with autism spectrum disorder (ASD). Nevertheless, specific cellular and molecular mechanisms underlying these alterations remain to be elucidated. In the present Editorial, we highlight an article in the current issue of the Journal of Neurochemistry that provides first evidence for the structural and cellular basis of an atypical corpus callosum long-distance connectivity impairments observed in ASD patients. The authors used a juvenile valproic acid (VPA) rat model of ASD that presents with reduced myelin level, specifically in the corpus callosum, and with an altered myelin sheet structure that is closely associated with the behavioral alteration found in these rats. This hypomyelination occurs primarily during infancy prior to oligodendroglial alterations, implicating that axonal-oligodendroglial connections are compromised in this model. Concomitant with the hypomyelination, the ASD rat model showed an atypical brain metabolic pattern, with hypometabolic activity across the whole brain, and hypermetabolism in brain areas related to autistic-like behavior. These findings contribute to unravel the neurobiological basis underlying white matter alteration and altered long-distance brain connectivity as described in ASD, paving the way to the development of new early diagnostic markers and toward developing future specific therapies for ASD.


Subject(s)
Autistic Disorder/chemically induced , Autistic Disorder/metabolism , Corpus Callosum/metabolism , Nerve Net/metabolism , Valproic Acid/toxicity , Animals , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Autistic Disorder/pathology , Brain/metabolism , Brain/pathology , Corpus Callosum/drug effects , Humans , Nerve Net/drug effects , Nerve Net/pathology , Rats
13.
Mol Psychiatry ; 26(12): 7596-7609, 2021 12.
Article in English | MEDLINE | ID: mdl-34331007

ABSTRACT

Shank3 monogenic mutations lead to autism spectrum disorders (ASD). Shank3 is part of the glutamate receptosome that physically links ionotropic NMDA receptors to metabotropic mGlu5 receptors through interactions with scaffolding proteins PSD95-GKAP-Shank3-Homer. A main physiological function of the glutamate receptosome is to control NMDA synaptic function that is required for plasticity induction. Intact glutamate receptosome supports glutamate receptors activation and plasticity induction, while glutamate receptosome disruption blocks receptors activity, preventing the induction of subsequent plasticity. Despite possible impact on metaplasticity and cognitive behaviors, scaffold interaction dynamics and their consequences are poorly defined. Here, we used mGlu5-Homer interaction as a biosensor of glutamate receptosome integrity to report changes in synapse availability for plasticity induction. Combining BRET imaging and electrophysiology, we show that a transient neuronal depolarization inducing NMDA-dependent plasticity disrupts glutamate receptosome in a long-lasting manner at synapses and activates signaling pathways required for the expression of the initiated neuronal plasticity, such as ERK and mTOR pathways. Glutamate receptosome disruption also decreases the NMDA/AMPA ratio, freezing the sensitivity of the synapse to subsequent changes of neuronal activity. These data show the importance of a fine-tuning of protein-protein interactions within glutamate receptosome, driven by changes of neuronal activity, to control plasticity. In a mouse model of ASD, a truncated mutant form of Shank3 prevents the integrity of the glutamate receptosome. These mice display altered plasticity, anxiety-like, and stereotyped behaviors. Interestingly, repairing the integrity of glutamate receptosome and its sensitivity to the neuronal activity rescued synaptic transmission, plasticity, and some behavioral traits of Shank3∆C mice. Altogether, our findings characterize mechanisms by which Shank3 mutations cause ASD and highlight scaffold dynamics as new therapeutic target.


Subject(s)
Autistic Disorder , Microfilament Proteins , Nerve Tissue Proteins , Animals , Autistic Disorder/genetics , Autistic Disorder/metabolism , Disease Models, Animal , Endosomes/metabolism , Glutamic Acid/metabolism , Mice , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Synapses/metabolism
14.
Brain Res Bull ; 172: 229-235, 2021 07.
Article in English | MEDLINE | ID: mdl-33964347

ABSTRACT

Epilepsy is one of the most prevalent serious brain disorders worldwide. Accumulating evidence has suggested that inflammation participates in the progression and pathogenesis of epilepsy. During inflammation, a cytosolic multimolecular complex called the "inflammasome" is activated, driving the innate immune response. This inflammatory pathway by sensing various pathogens and molecules from damaged cells and then activation of caspase-1 enzyme initiates inflammatory responses. Activated caspase-1 leads to the proteolytic cleavage of the pro-inflammatory cytokines, interleukin-1ß (IL-1ß) and interleukin-18 (IL-18), and also induction of an inflammatory programmed cell death termed pyroptosis. NLR family pyrin domain-containing 1 (NLRP1) and NLRP3 are the two best-characterized inflammasome members, and both basic and clinical research has reported their activation during epilepsy. This overview is intended to summarize the current literature concerning NLRP1 and NLRP3 inflammasome activation and epilepsy.


Subject(s)
Epilepsy/metabolism , Inflammasomes/metabolism , Seizures/metabolism , Animals , Cell Death/physiology , Humans , Inflammation/metabolism , Status Epilepticus/metabolism
16.
Curr Opin Pharmacol ; 56: 93-101, 2021 02.
Article in English | MEDLINE | ID: mdl-33429227

ABSTRACT

The N-methyl-d-aspartate (NMDA) receptor, among the ionotropic glutamate receptors, are fundamental to integrating and transducing complex signaling in neurons. Glutamate activation of these receptors mediates intracellular signals essential to neuronal and synaptic formation and synaptic plasticity and also contribute to excitotoxic processes in several neurological disorders. The NMDA receptor signaling is mediated by the permeability to Ca2+ and by the large network of signaling and scaffolding proteins associated mostly with the large C-terminal domain of GluN2 subunits. Important studies showed that GluN2 C-terminal interactions differ in accordance with the GluN2 subtype, and this influences the type of signaling that NMDA receptor activity controls. Thus, it is not surprising that mutations in genes that codify for NMDA receptor subunits have been associated with severe neuronal diseases. We will review recent advances and explore outstanding problems in this active area of research.


Subject(s)
Neurons , Receptors, N-Methyl-D-Aspartate , Humans , Neuronal Plasticity , Neurons/metabolism , Protein Subunits/metabolism , Signal Transduction
17.
EMBO J ; 40(5): e104267, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33491217

ABSTRACT

Impairments in social relationships and awareness are features observed in autism spectrum disorders (ASDs). However, the underlying mechanisms remain poorly understood. Shank2 is a high-confidence ASD candidate gene and localizes primarily to postsynaptic densities (PSDs) of excitatory synapses in the central nervous system (CNS). We show here that loss of Shank2 in mice leads to a lack of social attachment and bonding behavior towards pubs independent of hormonal, cognitive, or sensitive deficits. Shank2-/- mice display functional changes in nuclei of the social attachment circuit that were most prominent in the medial preoptic area (MPOA) of the hypothalamus. Selective enhancement of MPOA activity by DREADD technology re-established social bonding behavior in Shank2-/- mice, providing evidence that the identified circuit might be crucial for explaining how social deficits in ASD can arise.


Subject(s)
Autistic Disorder/drug therapy , Disease Models, Animal , Interpersonal Relations , Maternal Behavior/drug effects , Nerve Tissue Proteins/physiology , Piperazines/pharmacology , Preoptic Area/drug effects , Animals , Autistic Disorder/etiology , Autistic Disorder/metabolism , Autistic Disorder/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Preoptic Area/metabolism , Preoptic Area/pathology , Synapses
18.
Mol Psychiatry ; 26(6): 1928-1944, 2021 06.
Article in English | MEDLINE | ID: mdl-33402706

ABSTRACT

Human mutations and haploinsufficiency of the SHANK family genes are associated with autism spectrum disorders (ASD) and intellectual disability (ID). Complex phenotypes have been also described in all mouse models of Shank mutations and deletions, consistent with the heterogeneity of the human phenotypes. However, the specific role of Shank proteins in synapse and neuronal functions remain to be elucidated. Here, we generated a new mouse model to investigate how simultaneously deletion of Shank1 and Shank3 affects brain development and behavior in mice. Shank1-Shank3 DKO mice showed a low survival rate, a developmental strong reduction in the activation of intracellular signaling pathways involving Akt, S6, ERK1/2, and eEF2 during development and a severe behavioral impairments. Our study suggests that Shank1 and Shank3 proteins are essential to developmentally regulate the activation of Akt and correlated intracellular pathways crucial for mammalian postnatal brain development and synaptic plasticity. Therefore, Akt function might represent a new therapeutic target for enhancing cognitive abilities of syndromic ASD patients.


Subject(s)
Autism Spectrum Disorder , Proto-Oncogene Proteins c-akt , Animals , Autism Spectrum Disorder/genetics , Humans , Mice , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/genetics , Synapses
19.
Nat Commun ; 11(1): 6194, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33273479

ABSTRACT

Genetic mosaicism, a condition in which an organ includes cells with different genotypes, is frequently present in monogenic diseases of the central nervous system caused by the random inactivation of the X-chromosome, in the case of X-linked pathologies, or by somatic mutations affecting a subset of neurons. The comprehension of the mechanisms of these diseases and of the cell-autonomous effects of specific mutations requires the generation of sparse mosaic models, in which the genotype of each neuron is univocally identified by the expression of a fluorescent protein in vivo. Here, we show a dual-color reporter system that, when expressed in a floxed mouse line for a target gene, leads to the creation of mosaics with tunable degree. We demonstrate the generation of a knockout mosaic of the autism/epilepsy related gene PTEN in which the genotype of each neuron is reliably identified, and the neuronal phenotype is accurately characterized by two-photon microscopy.


Subject(s)
Fluorescent Dyes/chemistry , Genes, Reporter , Integrases/metabolism , Mosaicism , Neurodevelopmental Disorders/genetics , Action Potentials , Animals , Animals, Newborn , Disease Models, Animal , Electroencephalography , Gene Expression , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neurodevelopmental Disorders/physiopathology , PTEN Phosphohydrolase/metabolism , Tamoxifen/pharmacology
20.
Ther Adv Psychopharmacol ; 10: 2045125320968331, 2020.
Article in English | MEDLINE | ID: mdl-33224469

ABSTRACT

Approximately 30% of Major Depressive Disorder (MDD) patients develop treatment-resistant depression (TRD). Among the different causes that make TRD so challenging in both clinical and research contexts, major roles are played by the inadequate understanding of MDD pathophysiology and the limitations of current pharmacological treatments. Nevertheless, the field of psychiatry is facing exciting times. Combined with recent advances in genome editing techniques, human induced pluripotent stem cell (hiPSC) technology is offering novel and unique opportunities in both disease modelling and drug discovery. This technology has allowed innovative disease-relevant patient-specific in vitro models to be set up for many psychiatric disorders. Such models hold great potential in enhancing our understanding of MDD pathophysiology and overcoming many of the well-known practical limitations inherent to animal and post-mortem models. Moreover, the field is approaching the advent of (es)ketamine, a glutamate N-methyl-d-aspartate (NMDA) receptor antagonist, claimed as one of the first and exemplary agents with rapid (in hours) antidepressant effects, even in TRD patients. Although ketamine seems poised to transform the treatment of depression, its exact mechanisms of action are still unclear but greatly demanded, as the resulting knowledge may provide a model to understand the mechanisms behind rapid-acting antidepressants, which may lead to the discovery of novel compounds for the treatment of depression. After reviewing insights into ketamine's mechanisms of action (derived from preclinical animal studies) and depicting the current state of the art of hiPSC technology below, we will consider the implementation of an hiPSC technology-based TRD model for the study of ketamine's fast acting antidepressant mechanisms of action.

SELECTION OF CITATIONS
SEARCH DETAIL
...