← Back

Mouse Model

Topic spotlight
TopicWorld Wide

mouse model

Discover seminars, jobs, and research tagged with mouse model across World Wide.
100 curated items60 Seminars40 ePosters
Updated about 1 month ago
100 items · mouse model
100 results
SeminarNeuroscience

MRI investigation of orientation-dependent changes in microstructure and function in a mouse model of mild traumatic brain injury

Amr Eed
Western University
Nov 5, 2025
SeminarNeuroscience

Neural mechanisms of optimal performance

Luca Mazzucato
University of Oregon
May 22, 2025

When we attend a demanding task, our performance is poor at low arousal (when drowsy) or high arousal (when anxious), but we achieve optimal performance at intermediate arousal. This celebrated Yerkes-Dodson inverted-U law relating performance and arousal is colloquially referred to as being "in the zone." In this talk, I will elucidate the behavioral and neural mechanisms linking arousal and performance under the Yerkes-Dodson law in a mouse model. During decision-making tasks, mice express an array of discrete strategies, whereby the optimal strategy occurs at intermediate arousal, measured by pupil, consistent with the inverted-U law. Population recordings from the auditory cortex (A1) further revealed that sound encoding is optimal at intermediate arousal. To explain the computational principle underlying this inverted-U law, we modeled the A1 circuit as a spiking network with excitatory/inhibitory clusters, based on the observed functional clusters in A1. Arousal induced a transition from a multi-attractor (low arousal) to a single attractor phase (high arousal), and performance is optimized at the transition point. The model also predicts stimulus- and arousal-induced modulations of neural variability, which we confirmed in the data. Our theory suggests that a single unifying dynamical principle, phase transitions in metastable dynamics, underlies both the inverted-U law of optimal performance and state-dependent modulations of neural variability.

SeminarNeuroscience

Influence of the context of administration in the antidepressant-like effects of the psychedelic 5-MeO-DMT

Romain Hacquet
Université de Toulouse
Aug 28, 2024

Psychedelics like psilocybin have shown rapid and long-lasting efficacy on depressive and anxiety symptoms. Other psychedelics with shorter half-lives, such as DMT and 5-MeO-DMT, have also shown promising preliminary outcomes in major depression, making them interesting candidates for clinical practice. Despite several promising clinical studies, the influence of the context on therapeutic responses or adverse effects remains poorly documented. To address this, we conducted preclinical studies evaluating the psychopharmacological profile of 5-MeO-DMT in contexts previously validated in mice as either pleasant (positive setting) or aversive (negative setting). Healthy C57BL/6J male mice received a single intraperitoneal (i.p.) injection of 5-MeO-DMT at doses of 0.5, 5, and 10 mg/kg, with assessments at 2 hours, 24 hours, and one week post-administration. In a corticosterone (CORT) mouse model of depression, 5-MeO-DMT was administered in different settings, and behavioral tests mimicking core symptoms of depression and anxiety were conducted. In CORT-exposed mice, an acute dose of 0.5 mg/kg administered in a neutral setting produced antidepressant-like effects at 24 hours, as observed by reduced immobility time in the Tail Suspension Test (TST). In a positive setting, the drug also reduced latency to first immobility and total immobility time in the TST. However, these beneficial effects were negated in a negative setting, where 5-MeO-DMT failed to produce antidepressant-like effects and instead elicited an anxiogenic response in the Elevated Plus Maze (EPM).Our results indicate a strong influence of setting on the psychopharmacological profile of 5-MeO-DMT. Future experiments will examine cortical markers of pre- and post-synaptic density to correlate neuroplasticity changes with the behavioral effects of 5-MeO-DMT in different settings.

SeminarNeuroscience

Investigating dynamiCa++l mechanisms underlying cortical development and disease

Georgia Panagiotakos
Icahn School of Medicine at Mount Sinai
May 7, 2024
SeminarNeuroscience

Investigating activity-dependent processes during cortical neuronal assembly in development and disease

Simona Lodato
Humanitas University
Mar 19, 2024
SeminarNeuroscience

Astrocyte reprogramming / activation and brain homeostasis

Thomaidou Dimitra
Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
Dec 12, 2023

Astrocytes are multifunctional glial cells, implicated in neurogenesis and synaptogenesis, supporting and fine-tuning neuronal activity and maintaining brain homeostasis by controlling blood-brain barrier permeability. During the last years a number of studies have shown that astrocytes can also be converted into neurons if they force-express neurogenic transcription factors or miRNAs. Direct astrocytic reprogramming to induced-neurons (iNs) is a powerful approach for manipulating cell fate, as it takes advantage of the intrinsic neural stem cell (NSC) potential of brain resident reactive astrocytes. To this end, astrocytic cell fate conversion to iNs has been well-established in vitro and in vivo using combinations of transcription factors (TFs) or chemical cocktails. Challenging the expression of lineage-specific TFs is accompanied by changes in the expression of miRNAs, that post-transcriptionally modulate high numbers of neurogenesis-promoting factors and have therefore been introduced, supplementary or alternatively to TFs, to instruct direct neuronal reprogramming. The neurogenic miRNA miR-124 has been employed in direct reprogramming protocols supplementary to neurogenic TFs and other miRNAs to enhance direct neurogenic conversion by suppressing multiple non-neuronal targets. In our group we aimed to investigate whether miR-124 is sufficient to drive direct reprogramming of astrocytes to induced-neurons (iNs) on its own both in vitro and in vivo and elucidate its independent mechanism of reprogramming action. Our in vitro data indicate that miR-124 is a potent driver of the reprogramming switch of astrocytes towards an immature neuronal fate. Elucidation of the molecular pathways being triggered by miR-124 by RNA-seq analysis revealed that miR-124 is sufficient to instruct reprogramming of cortical astrocytes to immature induced-neurons (iNs) in vitro by down-regulating genes with important regulatory roles in astrocytic function. Among these, the RNA binding protein Zfp36l1, implicated in ARE-mediated mRNA decay, was found to be a direct target of miR-124, that be its turn targets neuronal-specific proteins participating in cortical development, which get de-repressed in miR-124-iNs. Furthermore, miR-124 is potent to guide direct neuronal reprogramming of reactive astrocytes to iNs of cortical identity following cortical trauma, a novel finding confirming its robust reprogramming action within the cortical microenvironment under neuroinflammatory conditions. In parallel to their reprogramming properties, astrocytes also participate in the maintenance of blood-brain barrier integrity, which ensures the physiological functioning of the central nervous system and gets affected contributing to the pathology of several neurodegenerative diseases. To study in real time the dynamic physical interactions of astrocytes with brain vasculature under homeostatic and pathological conditions, we performed 2-photon brain intravital imaging in a mouse model of systemic neuroinflammation, known to trigger astrogliosis and microgliosis and to evoke changes in astrocytic contact with brain vasculature. Our in vivo findings indicate that following neuroinflammation the endfeet of activated perivascular astrocytes lose their close proximity and physiological cross-talk with vasculature, however this event is at compensated by the cross-talk of astrocytes with activated microglia, safeguarding blood vessel coverage and maintenance of blood-brain integrity.

SeminarNeuroscienceRecording

Circadian modulation by time-restricted feeding rescues brain pathology and improves memory in mouse models of Alzheimer’s disease

Daniel S. Whittaker
UCSD
Nov 8, 2023
SeminarNeuroscience

X-linked mosaicism and behavioral heterogeneity in Rett syndrome

Keerthi Krishnan
University of Tennessee, Knoxville
Oct 31, 2023
SeminarNeuroscience

Cellular crosstalk in Neurodevelopmental Disorders

Silvia Cappello
Max Planck Institute
Sep 26, 2023

Cellular crosstalk is an essential process during brain development and it is influenced by numerous factors, including the morphology of the cells, their adhesion molecules, the local extracellular matrix and the secreted vesicles. Inspired by mutations associated with neurodevelopmental disorders, we focus on understanding the role of extracellular mechanisms essential for the correct development of the human brain. Hence, we combine the in vivo mouse model and the in vitro human-derived neurons, cerebral organoids, and dorso-ventral assembloids in order to better comprehend the molecular and cellular mechanisms involved in ventral progenitors’ proliferation and fate as well as migration and maturation of inhibitory neurons during human brain development and tackle the causes of neurodevelopmental disorders. We particularly focus on mutations in genes influencing cell-cell contacts, extracellular matrix, and secretion of vesicles and therefore study intrinsic and extrinsic mechanisms contributing to the formation of the brain. Our data reveal an important contribution of cell non-autonomous mechanisms in the development of neurodevelopmental disorders.

SeminarNeuroscience

Circuit mechanisms of attention dysfunction in Scn8a+/- mice: implications for epilepsy and neurodevelopmental disorders

Brielle Ferguson
Harvard Medical School
May 16, 2023
SeminarNeuroscienceRecording

Perivascular cells induce microglial phagocytic states and synaptic engulfment via SPP1 in mouse models of Alzheimer's disease

Sebastiaan de Schepper
University College London
May 15, 2023
SeminarNeuroscienceRecording

Brain mosaicism in epileptogenic cortical malformations

Stéphanie Baulac
ICM Paris
Jan 31, 2023

Focal Cortical Dysplasia (FCD) is the most common focal cortical malformation leading to intractable childhood focal epilepsy. In recent years, we and others have shown that FCD type II is caused by mosaic mutations in genes within the PI3K-AKT-mTOR-signaling pathway. Hyperactivation of the mTOR pathway accounts for neuropathological abnormalities and seizure occurrence in FCD. We further showed from human surgical FCDII tissue that epileptiform activity correlates with the density of mutated dysmorphic neurons, supporting their pro-epileptogenic role. The level of mosaicism, as defined by variant allele frequency (VAF) is thought to correlate with the size and regional brain distribution of the lesion such that when a somatic mutation occurs early during the cortical development, the dysplastic area is smaller than if it occurs later. Novel approaches based on the detection of cell-free DNA from the CSF and from trace tissue adherent to SEEG electrodes promise future opportunities for genetic testing during the presurgical evaluation of refractory epilepsy patients or in those that are not eligible for surgery. In utero-based electroporation mouse models allow to express somatic mutation during neurodevelopment and recapitulate most neuropathological and clinical features of FCDII, establishing relevant preclinical mouse models for developing precision medicine strategies.

SeminarNeuroscience

How do Astrocytes Sculpt Synaptic Circuits?

Cagla Eroglu
Duke University
Jan 10, 2023
SeminarNeuroscience

From symptoms to circuits in Fragile X syndrome

Carlos Portera-Cailliau
University of California, Los Angeles
Dec 20, 2022
SeminarNeuroscienceRecording

Protective microglial signaling in Alzheimer's Disease

Hannah Ennerfelt
Stanford University
Dec 15, 2022

Recent studies have begun to reveal critical roles for the brain’s professional phagocytes, microglia, and their receptors in the control of neurotoxic amyloid beta (Aβ) and myelin debris accumulation in neurodegenerative disease. However, the critical intracellular molecules that orchestrate neuroprotective functions of microglia remain poorly understood. In our studies, we find that targeted deletion of SYK in microglia leads to exacerbated Aβ deposition, aggravated neuropathology, and cognitive defects in the 5xFAD mouse model of Alzheimer’s disease (AD). Disruption of SYK signaling in this AD model was further shown to impede the development of disease-associated microglia (DAM), alter AKT/GSK3β-signaling, and restrict Aβ phagocytosis by microglia. Conversely, receptor-mediated activation of SYK limits Aβ load. We also found that SYK critically regulates microglial phagocytosis and DAM acquisition in demyelinating disease. Collectively, these results broaden our understanding of the key innate immune signaling molecules that instruct beneficial microglial functions in response to neurotoxic material." https://doi.org/10.1016/j.cell.2022.09.030

SeminarNeuroscience

Dysregulated Translation in Fragile X Syndrome

Eric Klann
New York University
Nov 8, 2022
SeminarNeuroscienceRecording

Hypothalamic episode generators underlying the neural control of fertility

Allan Herbison
Department of Physiology, Development and Neuroscience, University of Cambridge
Nov 7, 2022

The hypothalamus controls diverse homeostatic functions including fertility. Neural episode generators are required to drive the intermittent pulsatile and surge profiles of reproductive hormone secretion that control gonadal function. Studies in genetic mouse models have been fundamental in defining the neural circuits forming these central pattern generators and the full range of in vitro and in vivo optogenetic and chemogenetic methodologies have enabled investigation into their mechanism of action. The seminar will outline studies defining the hypothalamic “GnRH pulse generator network” and current understanding of its operation to drive pulsatile hormone secretion.

SeminarNeuroscience

Counteracting epigenetic mechanisms in autism spectrum disorders

Sofia Lizarraga
University of South Carolina
Oct 11, 2022
SeminarNeuroscience

SCN8A (Nav1.6) and DEE:  mouse models and pre-clinical therapies

Miriam Meisler
University of Michigan
Sep 6, 2022

SCN8A encodes a major voltage-gated sodium channel expressed in CNS and PNS neurons.  Gain-of-function and loss-of-function mutations contribute to  human disorders, most notably Developmental and Epileptic Encephalophy (DEE). More than 600 affected individuals have been reported, with the most common  mechanism of de novo, gain-of-function mutations.  We have developed constitutive  and conditional models of gain- and loss- of function mutations in the mouse and  characterized the effects of on neuronal firing and neurological phenotypes.  Using CRE lines with cellular and developmental specificity, we have probed the effects of activating  mutant alleles in various classes of neurons in the developing and adult mouse.   Most recently, we are testing genetic therapies that reduce the expression  of gain-of-function mutant alleles.  We are comparing the effectiveness of allele specific  oligos (ASOs), viral delivery of shRNAs, and allele-specific targeting of mutant alleles  using Crispr/Cas9 in mouse models of DEE.

SeminarNeuroscience

Epigenome regulation in neocortex expansion and generation of neuronal subtypes

Tran Tuoc, PhD
Ruhruniversität-Bochum, Humangenetik
Aug 23, 2022

Evolutionarily, the expansion of the human neocortex accounts for many of the unique cognitive abilities of humans. This expansion appears to reflect the increased proliferative potential of basal progenitors (BPs) in mammalian evolution. Further cortical progenitors generate both glutamatergic excitatory neurons (ENs) and GABAergic inhibitory interneurons (INs) in human cortex, whereas they produce exclusively ENs in rodents. The increased proliferative capacity and neuronal subtype generation of cortical progenitors in mammalian evolution may have evolved through epigenetic alterations. However, whether or how the epigenome in cortical progenitors differs between humans and other species is unknown. Here, we report that histone H3 acetylation is a key epigenetic regulation in BP profiling of sorted BPs, we show that H3K9 acetylation is low in murine BPs and high in amplification, neuronal subtype generation and cortical expansion. Through epigenetic profiling of sorted BPs, we show that H3K9 acetylation is low in murine BPs and high in human BPs. Elevated H3K9ac preferentially increases BP proliferation, increasing the size and folding of the normally smooth mouse neocortex. Furthermore, we found that the elevated H3 acetylation activates expression of IN genes in in developing mouse cortex and promote proliferation of IN progenitor-like cells in cortex of Pax6 mutant mouse models. Mechanistically, H3K9ac drives the BP amplification and proliferation of these IN progenitor-like cells by increasing expression of the evolutionarily regulated gene, TRNP1. Our findings demonstrate a previously unknown mechanism that controls neocortex expansion and generation of neuronal subtypes. Keywords: Cortical development, neurogenesis, basal progenitors, cortical size, gyrification, excitatory neuron, inhibitory interneuron, epigenetic profiling, epigenetic regulation, H3 acetylation, H3K9ac, TRNP1, PAX6

SeminarNeuroscience

Role of ASD risk genes on maturation of frontal-sensory cognitive control circuit

Hiro Morishita
Icahn School of Medicine at Mount Sinai
Jul 26, 2022
SeminarNeuroscience

At the nexus of genes, aging and environment: Understanding transcriptomic and epigenomic regulation in Parkinson's disease

Julia Schulze-Hentrich
Institute of Medical Genetics and Applied Genomics, University of Tübingen
Jul 19, 2022

Parkinson’s Disease (PD), the most common neurodegenerative movement disorder, is based on a complex interplay between genetic predispositions, aging processes, and environmental influences. In order to better understand the gene-environment axis in PD, we pursue a multi-omics approach to comprehensively interrogate genome-wide changes in histone modifications, DNA methylation, and hydroxymethylation, accompanied by transcriptomic profiling in cell and animal models of PD as well as large patient cohorts. Furthermore, we assess the plasticity of epigenomic modifications under influence of environmental factors using longitudinal cohorts of sporadic PD cases as well as mouse models exposed to specific environmental factors. Here, we present gene expression changes in PD mouse models in context of aging as well as environmental enrichment and high-fat diet.

SeminarNeuroscience

Elucidating the mechanism underlying Stress and Caffeine-induced motor dysfunction using a mouse model of Episodic Ataxia Type 2

Heather Snell
Albert Einstein Medical College
Apr 26, 2022

Episodic Ataxia type 2 (EA2), caused by mutations in the CACNA1A gene, results in a loss-of-function of the P/Q type calcium channel, which leads to baseline ataxia, and attacks of dyskinesia, that can last a few hours to a few days. Attacks are brought on by consumption of caffeine, alcohol, and physical or emotional stress. Interestingly, caffeine and stress are common triggers among other episodic channelopathies, as well as causing tremor or shaking in otherwise healthy adults. The mechanism underlying stress and caffeine induced motor impairment remains poorly understood. Utilizing behavior, and in vivo and in vitro electrophysiology in the tottering mouse, a well characterized mouse model of EA2, or WT mice, we first sought to elucidate the mechanism underlying stress-induced motor impairment. We found stress induces attacks in EA2 though the activation of cerebellar alpha 1 adrenergic receptors by norepinephrine (NE) through casein kinase 2 (CK2) dependent phosphorylation. This decreases SK2 channel activity, causing increased Purkinje cell irregularity and motor impairment. Knocking down or blocking CK2 with an FDA approved drug CX-4945 prevented PC irregularity and stress-induced attacks. We next hypothesized caffeine, which has been shown to increase NE levels, could induce attacks through the same alpha 1 adrenergic mechanism in EA2. We found caffeine increases PC irregularity and induces attacks through the same CK2 pathway. Block of alpha 1 adrenergic receptors, however, failed to prevent caffeine-induced attacks. Caffeine instead induces attacks through the block of cerebellar A1 adenosine receptors. This increases the release of glutamate, which interacts with mGluR1 receptors on PC, resulting in erratic firing and motor attacks. Finally, we show a novel direct interaction between mGluR1 and CK2, and inhibition of mGluR1 prior to initiation of attack, prevents the caffeine-induced increase in phosphorylation. These data elucidate the mechanism underlying stress and caffeine-induced motor impairment. Furthermore, given the success of CX-4945 to prevent stress and caffeine induced attacks, it establishes ground-work for the development of therapeutics for the treatment of caffeine and stress induced attacks in EA2 patients and possibly other episodic channelopathies.

SeminarNeuroscience

Memory, learning to learn, and control of cognitive representations

Andre Fenton
New York University
Apr 19, 2022
SeminarNeuroscience

Studying cortical development through the lens of autism spectrum disorders

Gaia Novarino
Institute of Science and Technology Austria
Feb 22, 2022
SeminarNeuroscience

Primary Motor Cortex Circuitry in a Mouse Model of Parkinson’s Disease

Olivia Swanson
Dani lab, University of Pennsylvania
Feb 8, 2022

The primary motor cortex (M1) is a major output center for movement execution and motor learning, and its dysfunction contributes to the pathophysiology of Parkinson’s disease (PD). While human studies have indicated that a loss of midbrain dopamine neurons alters M1 activation, the mechanisms underlying this phenomenon remain unclear. Using a mouse model of PD, we uncovered several shifts within M1 circuitry following dopamine depletion, including impaired excitation by thalamocortical afferents and altered excitability. Our findings add to the growing body of literature highlighting M1 as a major contributor in PD, and provide targeted neural substrates for possible therapeutic interventions.

SeminarNeuroscience

Reward system function and dysfunction in Autism Spectrum Disorders

Camilla Bellone
University of Geneva
Feb 8, 2022
SeminarNeuroscienceRecording

Genetics of migraine and the use of genetic mouse models

Arn van den Maagdenberg
Departments of Human Genetics and Neurology, Leiden University Medical Centre, Leiden, the Netherlands
Jan 26, 2022
SeminarNeuroscience

Translational Biomarkers in Preclinical Models of Neurodevelopmental Disorders

Jill Silverman
UC Davis
Jan 25, 2022
SeminarNeuroscience

Synaptic alterations in the striatum drive ASD-related behaviors in mice

Helen Bateup
UC Berkeley
Jan 11, 2022
SeminarNeuroscienceRecording

Mechanisms of sleep-seizure interactions in tuberous sclerosis and other mTORpathies

Michael Wong
Washigton University
Jan 4, 2022

An intriguing, relatively unexplored therapeutic avenue to investigate epilepsy is the interaction of sleep mechanisms and seizures. Multiple lines of clinical observations suggest a strong, bi-directional relationship between epilepsy and sleep. Epilepsy and sleep disorders are common comorbidities. Seizures occur more commonly in sleep in many types of epilepsy, and in turn, seizures can cause disrupted sleep. Sudden unexplained death in epilepsy (SUDEP) is strongly associated with sleep. The biological mechanisms underlying this relationship between seizures and sleep are poorly understood, but if better delineated, could offer novel therapeutic approaches to treating both epilepsy and sleep disorders. In this presentation, I will explore this sleep-seizure relationship in mouse models of epilepsy. First, I will present general approaches for performing detailed longitudinal sleep and vigilance state analysis in mice, including pre-weanling neonatal mice. I will then discuss recent data from my laboratory demonstrating an abnormal sleep phenotype in a mouse model of the genetic epilepsy, tuberous sclerosis complex (TSC), and its relationship to seizures. The potential mechanistic basis of sleep abnormalities and sleep-seizure interactions in this TSC model will be investigated, focusing on the role of the mechanistic target of rapamycin (mTOR) pathway and hypothalamic orexin, with potential therapeutic applications of mTOR inhibitors and orexin antagonists. Finally, similar sleep-seizure interactions and mechanisms will be extended to models of acquired epilepsy due to status epilepticus-related brain injury.

SeminarNeuroscience

The effects of maternal immune activation on early development in an outbred strain of mice

Tamara Franklin
Dalhousie University
Nov 23, 2021
SeminarNeuroscience

Stem cell approaches to understand acquired and genetic epilepsies

Jenny Hsieh
University of Texas at San Antonio
Nov 16, 2021

The Hsieh lab focuses on the mechanisms that promote neural stem cell self-renewal and differentiation in embryonic and adult brain. Using mouse models, video-EEG monitoring, viral techniques, and imaging/electrophysiological approaches, we elucidated many of the key transcriptional/epigenetic regulators of adult neurogenesis and showed aberrant new neuron integration in adult rodent hippocampus contribute to circuit disruption and seizure development. Building on this work, I will present our recent studies describing how GABA-mediated Ca2+ activity regulates the production of aberrant adult-born granule cells. In a new direction of my laboratory, we are using human induced pluripotent stem cells and brain organoid models as approaches to understand brain development and disease. Mutations in one gene, Aristaless-related homeobox (ARX), are of considerable interest since they are known to cause a common spectrum of neurodevelopmental disorders including epilepsy, autism, and intellectual disability. We have generated cortical and subpallial organoids from patients with poly-alanine expansion mutations in ARX. To understand the nature of ARX mutations in the organoid system, we are currently performing cellular, molecular, and physiological analyses. I will present these data to gain a comprehensive picture of the effect of ARX mutations in brain development. Since we do not understand how human brain development is affected by ARX mutations that contribute to epilepsy, we believe these studies will allow us to understand the mechanism of pathogenesis of ARX mutations, which has the potential to impact the diagnosis and care of patients.

SeminarNeuroscience

Gut-brain signaling as a driver of behavior and gene expression in a mouse model for autism spectrum disorder

Drew Kiraly
Icahn School of Medicine at Mount Sinai
Nov 9, 2021
SeminarNeuroscienceRecording

Mechanisms of CACNA1A-associated developmental epileptic encephalopathies

Elsa Rossignol
University of Montreal
Nov 2, 2021

Developmental epileptic encephalopathies are early-onset epilepsies, often refractory to therapy, with developmental delay or regression. These disorders carry poor neurodevelopmental prognosis, with long-term refractory epilepsy and persistent cognitive, behavioral and motor deficits. Mutations in the CACNA1A gene, encoding the pore-forming α1 subunit of CaV2.1 voltage-gated calcium channels, result in a spectrum of neurological disorders, including severe, early-onset epileptic encephalopathies. Recent work from the Rossignol lab helped characterize the phenotypic spectrum of CACNA1A-related epilepsies in humans. Using conditional genetics and novel animal models, the Rossignol lab unveiled some of the underlying pathophysiological mechanisms, including critical deficits in cortical inhibition, resulting in seizures and a range of cognitive-behavioral deficits. Importantly, Dr. Rossignol’s team demonstrated that the targeted activation of specific GABAergic interneuron populations in selected cortical regions prevents motor seizures and reverts attention deficits and cognitive rigidity in mouse models of the disorder. These recent findings open novel avenues for the treatment of these severe CACNA1A-associated neurodevelopmental disorders.

SeminarNeuroscienceRecording

Migraine: a disorder of excitatory-inhibitory balance in multiple brain networks? Insights from genetic mouse models of the disease

Daniela Pietrobon
Department of Biomedical Sciences and Padova Neuroscience Center, University of Padova, Italy
Oct 27, 2021

Migraine is much more than an episodic headache. It is a complex brain disorder, characterized by a global dysfunction in multisensory information processing and integration. In a third of patients, the headache is preceded by transient sensory disturbances (aura), whose neurophysiological correlate is cortical spreading depression (CSD). The molecular, cellular and circuit mechanisms of the primary brain dysfunctions that underlie migraine onset, susceptibility to CSD and altered sensory processing remain largely unknown and are major open issues in the neurobiology of migraine. Genetic mouse models of a rare monogenic form of migraine with aura provide a unique experimental system to tackle these key unanswered questions. I will describe the functional alterations we have uncovered in the cerebral cortex of genetic mouse models and discuss the insights into the cellular and circuit mechanisms of migraine obtained from these findings.

SeminarNeuroscience

Evidence for the role of glymphatic dysfunction in the development of Alzheimer’s disease

Jeffrey Iliff
VA Puget Sound Health Care System, University of Washignton, Seattle, WA, USA
Oct 24, 2021

Glymphatic perivascular exchange is supported by the astroglial water channel aquaporin-4 (AQP4), which localizes to perivascular astrocytic endfeet surrounding the cerebral vasculature. In aging mice, impairment of glymphatic function is associated with reduced perivascular AQP4 localization, yet whether these changes contribute to the development of neurodegenerative disease, such as Alzheimer’s disease (AD), remains unknown. Using post mortem human tissue, we evaluated perivascular AQP4 localization in the frontal cortical gray matter, white matter, and hippocampus of cognitively normal subjects and those with AD. Loss of perivascular and increasing cellular localization of AQP4 in the frontal gray matter was specifically associated with AD status, amyloid β (Aβ) and tau pathology, and cognitive decline in the early stages of disease. Using AAV-PHP.B to drive expression on non-perivascular AQP4 in wild type and Tg2576 (APPSwe, mouse model of Aβ deposition) mice, increased cellular AQP4 localization did not slow glymphatic function or change Aβ deposition. Using the Snta1 knockout line (which lacks perivascular AQP4 localization), we observed that loss AQP4 from perivascular endfeet slowed glymphatic function in wild type mice and accelerated Aβ plaque deposition in Tg2576 mice. These findings demonstrate that loss of perivascular AQP4 localization, and not increased cellular AQP4 localization, slows glymphatic function and promotes the development of AD pathology. To evaluate whether naturally occurring variation in the human AQP4 gene, or the alpha syntrophin (SNTA1), dystrobrevin (DTNA) or dystroglycan (DAG1) genes (whose products maintain perivascular AQP4 localization) confer risk for or protection from AD pathology or clinical progression, we evaluated 56 tag single nucleotide polymorphisms (SNPs) across these genes for association with CSF AD biomarkers, MRI measures of cortical and hippocampal atrophy, and longitudinal cognitive decline in the Alzheimer’s Disease Neuroimaging Initiative I (ADNI I) cohort. We identify 25 different significant associations between AQP4, SNTA1, DTNA, and DAG1 tag SNPs and phenotypic measures of AD pathology and progression. These findings provide complimentary human genetic evidence for the contribution of perivascular glymphatic dysfunction to the development of AD in human populations.

SeminarNeuroscienceRecording

Dancing to a Different Tune: TANGO Gives Hope for Dravet Syndrome

Lori Isom
University of Michigan
Oct 19, 2021

The long-term goal of our research is to understand the mechanisms of SUDEP, defined as Sudden, Unexpected, witnessed or unwitnessed, nontraumatic and non-drowning Death in patients with EPilepsy, excluding cases of documented status epilepticus. The majority of SUDEP patients die during sleep. SUDEP is the most devastating consequence of epilepsy, yet little is understood about its causes and no biomarkers exist to identify at risk patients. While SUDEP accounts for 7.5-20% of all epilepsy deaths, SUDEP risk in the genetic epilepsies varies with affected genes. Patients with ion channel gene variants have the highest SUDEP risk. Indirect evidence variably links SUDEP to seizure-induced apnea, pulmonary edema, dysregulation of cerebral circulation, autonomic dysfunction, and cardiac arrhythmias. Arrhythmias may be primary or secondary to hormonal or metabolic changes, or autonomic discharges. When SUDEP is compared to Sudden Cardiac Death secondary to Long QT Syndrome, especially to LQT3 linked to variants in the voltage-gated sodium channel (VGSC) gene SCN5A, there are parallels in the circumstances of death. To gain insight into SUDEP mechanisms, our approach has focused on channelopathies with high SUDEP incidence. One such disorder is Dravet syndrome (DS), a devastating form of developmental and epileptic encephalopathy (DEE) characterized by multiple pharmacoresistant seizure types, intellectual disability, ataxia, and increased mortality. While all patients with epilepsy are at risk for SUDEP, DS patients may have the highest risk, up to 20%, with a mean age at SUDEP of 4.6 years. Over 80% of DS is caused by de novo heterozygous loss-of-function (LOF) variants in SCN1A, encoding the VGSC Nav1.1  subunit, resulting in haploinsufficiency. A smaller cohort of patients with DS or a more severe DEE have inherited, homozygous LOF variants in SCN1B, encoding the VGSC 1/1B non-pore-forming subunits. A related DEE, Early Infantile EE (EIEE) type 13, is linked to de novo heterozygous gain-of-function variants in SCN8A, encoding the VGSC Nav1.6. VGSCs underlie the rising phase and propagation of action potentials in neurons and cardiac myocytes. SCN1A, SCN8A, and SCN1B are expressed in both the heart and brain of humans and mice. Because of this, we proposed that cardiac arrhythmias contribute to the mechanism of SUDEP in DEE. We have taken a novel approach to the development of therapeutics for DS in collaboration with Stoke Therapeutics. We employed Targeted Augmentation of Nuclear Gene Output (TANGO) technology, which modulates naturally occurring, non-productive splicing events to increase target gene and protein expression and ameliorate disease phenotype in a mouse model. We identified antisense oligonucleotides (ASOs) that specifically increase the expression of productive Scn1a transcript in human and mouse cell lines, as well as in mouse brain. We showed that a single intracerebroventricular dose of a lead ASO at postnatal day 2 or 14 reduced the incidence of electrographic seizures and SUDEP in the F1:129S-Scn1a+/- x C57BL/6J mouse model of DS. Increased expression of productive Scn1a transcript and NaV1.1 protein were confirmed in brains of treated mice. Our results suggest that TANGO may provide a unique, gene-specific approach for the treatment of DS.

SeminarNeuroscience

Behavioral phenotyping strategies for mouse models of neurodevelopmental disorders

Jacqueline N. Crawley
MIND Institute. Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
Sep 29, 2021
SeminarNeuroscience

On the role of the ADNP gene in mice and man

Frank Kooy
U Antwerpen
Sep 28, 2021
SeminarNeuroscienceRecording

Pitt-Hopkins Syndrome, mouse models, neurodevelopment, therapeutics

Andrew Kennedy
Bates College
Sep 14, 2021
SeminarNeuroscience

(Dys)regulation of the social brain

Claudia Bagni
Universite de Lausanne
Jun 22, 2021
SeminarNeuroscience

Developing a mouse incentive delay task

Miao Ge
Fudan University
Jun 22, 2021

Monetary incentive delay task (MID) is a well-validated human functional MRI task widely used in probing affective-motivational processes in psychiatric disorders. We are developing a mouse version of the MID task in order to facilitate translations of findings from the wealth of human imaging studies. This talk presents our task design and behavioural data from the ongoing work.

SeminarNeuroscienceRecording

The role of the complement pathway in post-traumatic sleep disruption and epilepsy

Jeanne Paz
UCSF
Jun 15, 2021

While traumatic brain injury (TBI) acutely disrupts the cortex, most TBI-related disabilities reflect secondary injuries that accrue over time. The thalamus is a likely site of secondary damage because of its reciprocal connections with the cortex. Using a mouse model of mild cortical injury that does not directly damage subcortical structures (mTBI), we found a chronic increase in C1q expression specifically in the corticothalamic circuit. Increased C1q expression co-localized with neuron loss and chronic inflammation, and correlated with disruption in sleep spindles and emergence of epileptic activities. Blocking C1q counteracted these outcomes, suggesting that C1q is a disease modifier in mTBI. Single-nucleus RNA sequencing demonstrated that microglia are the source of thalamic C1q. Since the corticothalamic circuit is important for cognition and sleep, which can be impaired by TBI, this circuit could be a new target for treating TBI-related disabilities

SeminarNeuroscience

Numbing intraneuronal Tau levels to prevent neurodegeneration in tauopathies

Michel Cayouette
Montreal Clinical Research Institute (IRCM)
May 30, 2021

Intraneuronal accumulation of the microtubule associated protein Tau is largely recognized as an important toxic factor linked to neuronal cell death in Alzheimer’s disease and tauopathies. While there has been progress uncovering mechanisms leading to the formation of toxic Tau tangles, less is known about how intraneuronal Tau levels are regulated in health and disease. Here, I will discuss our recent work showing that the intracellular trafficking adaptor protein Numb is critical to control intraneuronal Tau levels. Inactivation of Numb in retinal ganglion cells increases monomeric and oligomeric Tau levels and leads to axonal blebbing in optic nerves, followed by significant neuronal cell loss in old mice. Interestingly, overexpression of the long isoform of Numb (Numb-72) decreases intracellular Tau levels by promoting exocytosis of monomeric Tau. In TauP301S and triple transgenic AD mouse models, expression of Numb-72 in RGCs reduces the number of axonal blebs and prevents neurodegeneration. Finally, inactivation of Numb in TauP301S mice accelerates neurodegeneration in both the retina and spinal cord and leads to precocious paralysis. Taken together, these results uncover Numb as a essential regulator of Tau homeostasis in neurons and as a potential therapeutic agent for AD and tauopathies.

SeminarNeuroscienceRecording

A fresh look at the bird retina

Karin Dedek
University of Oldenburg
May 30, 2021

I am working on the vertebrate retina, with a main focus on the mouse and bird retina. Currently my work is focused on three major topics: Functional and molecular analysis of electrical synapses in the retina Circuitry and functional role of retinal interneurons: horizontal cells Circuitry for light-dependent magnetoreception in the bird retina Electrical synapses Electrical synapses (gap junctions) permit fast transmission of electrical signals and passage of metabolites by means of channels, which directly connect the cytoplasm of adjoining cells. A functional gap junction channel consists of two hemichannels (one provided by each of the cells), each comprised of a set of six protein subunits, termed connexins. These building blocks exist in a variety of different subtypes, and the connexin composition determines permeability and gating properties of a gap junction channel, thereby enabling electrical synapses to meet a diversity of physiological requirements. In the retina, various connexins are expressed in different cell types. We study the cellular distribution of different connexins as well as the modulation induced by transmitter action or change of ambient light levels, which leads to altered electrical coupling properties. We are also interested in exploiting them as therapeutic avenue for retinal degeneration diseases. Horizontal cells Horizontal cells receive excitatory input from photoreceptors and provide feedback inhibition to photoreceptors and feedforward inhibition to bipolar cells. Because of strong electrical coupling horizontal cells integrate the photoreceptor input over a wide area and are thought to contribute to the antagonistic organization of bipolar cell and ganglion cell receptive fields and to tune the photoreceptor–bipolar cell synapse with respect to the ambient light conditions. However, the extent to which this influence shapes retinal output is unclear, and we aim to elucidate the functional importance of horizontal cells for retinal signal processing by studying various transgenic mouse models. Retinal circuitry for light-dependent magnetoreception in the bird We are studying which neuronal cell types and pathways in the bird retina are involved in the processing of magnetic signals. Likely, magnetic information is detected in cryptochrome-expressing photoreceptors and leaves the retina through ganglion cell axons that project via the thalamofugal pathway to Cluster N, a part of the visual wulst essential for the avian magnetic compass. Thus, we aim to elucidate the synaptic connections and retinal signaling pathways from putatively magnetosensitive photoreceptors to thalamus-projecting ganglion cells in migratory birds using neuroanatomical and electrophysiological techniques.

SeminarNeuroscience

Towards targeted therapies for the treatment of Dravet Syndrome

Gaia Colasante
Ospedale San Raffaele
May 18, 2021

Dravet syndrome is a severe epileptic encephalopathy that begins during the first year of life and leads to severe cognitive and social interaction deficits. It is mostly caused by heterozygous loss-of-function mutations in the SCN1A gene, which encodes for the alpha-subunit of the voltage-gated sodium channel (Nav1.1) and is responsible mainly of GABAergic interneuron excitability. While different therapies based on the upregulation of the healthy allele of the gene are being developed, the dynamics of reversibility of the pathology are still unclear. In fact, whether and to which extent the pathology is reversible after symptom onset and if it is sufficient to ensure physiological levels of Scn1a during a specific critical period of time are open questions in the field and their answers are required for proper development of effective therapies. We generated a novel Scn1a conditional knock-in mouse model (Scn1aSTOP) in which the endogenous Scn1a gene is silenced by the insertion of a floxed STOP cassette in an intron of Scn1a gene; upon Cre recombinase expression, the STOP cassette is removed, and the mutant allele can be reconstituted as a functional Scn1a allele. In this model we can reactivate the expression of Scn1a exactly in the neuronal subtypes in which it is expressed and at its physiological level. Those aspects are crucial to obtain a final answer on the reversibility of DS after symptom onset. We exploited this model to demonstrate that global brain re-expression of the Scn1a gene when symptoms are already developed (P30) led to a complete rescue of both spontaneous and thermic inducible seizures and amelioration of behavioral abnormalities characteristic of this model. We also highlighted dramatic gene expression alterations associated with astrogliosis and inflammation that, accordingly, were rescued by Scn1a gene expression normalization at P30. Moreover, employing a conditional knock-out mouse model of DS we reported that ensuring physiological levels of Scn1a during the critical period of symptom appearance (until P30) is not sufficient to prevent the DS, conversely, mice start to die of SUDEP and develop spontaneous seizures. These results offer promising insights in the reversibility of DS and can help to accelerate therapeutic translation, providing important information on the timing for gene therapy delivery to Dravet patients.

SeminarNeuroscience

Dysregulation of mTOR Signaling Mediates Common Neurite and Migration Defects in Idiopathic and 16p11.2 Deletion Autism neural progenitors

Emanuel DiCicco-Bloom
Rutgers U
May 11, 2021
SeminarNeuroscience

Male-specific intracellular signaling and male bias in neurodevelopmental disorders

M Chiara Manzini
Rutgers U
Apr 27, 2021
SeminarNeuroscience

Targeting selective autophagy against neurodegenerative diseases

Ana Maria Cuervo
Albert Einstein College of Medicine, New York, USA
Apr 20, 2021

Protein quality control is essential for maintenance of a healthy and functional proteome that can attend the multiplicity of cellular functions. Failure of the systems that contribute to protein homeostasis, the so called proteostasis networks, have been identified in the pathogenesis of multiple neurodegenerative disorders and demonstrated to contribute to disease onset and progression. We are interested in autophagy, one of the components of the proteostasis network, and in the interplay of wo selective types of autophagy, chaperone-mediated autophagy (CMA) and endosomal microautophagy (eMI), with neurodegeneration. We have recently found that pathogenic proteins involved in common neurodegenerative conditions such as tauopathies or Parkinson’s disease, can exert a toxic effect in both types of selective types of autophagy compromising their functioning. We have now used mouse models with compromised CMA that support increased propagation of proteins such as tau and alpha-synuclein and an exacerbation of disease phenotype with aging. Conversely, genetic or chemical upregulation of CMA in this context of proteotoxicity slow down disease progression by facilitating effective intracellular removal of pathogenic proteins. Our findings highlight CMA and eMI as potential novel therapeutic targets against neurodegeneration.

SeminarNeuroscience

Sensory Processing and Arousal in Neurodevelopmental Disorders

Michela Fagiolini
Boston Children's Hospital
Apr 13, 2021
SeminarNeuroscience

Sex-Specific Brain Transcriptional Signatures in Human MDD and their Correlates in Mouse Models of Depression

Benoit Labonté
Université Laval & Centre de Recherche CERVO, Québec, Canada
Feb 11, 2021

Major depressive disorder (MDD) is a sexually dimorphic disease. This sexual dimorphism is believed to result from sex-specific molecular alterations affecting functional pathways regulating the capacity of men and women to cope with daily life stress differently. Transcriptional changes associated with epigenetic alterations have been observed in the brain of men and women with depression and similar changes have been reported in different animal models of stress-induced depressive-like behaviors. In fact, most of our knowledge of the biological basis of MDD is derived from studies of chronic stress models in rodents. However, while these models capture certain aspects of the features of MDD, the extent to which they reproduce the molecular pathology of the human syndrome remains unknown and the functional consequences of these changes on the neuronal networks controlling stress responses are poorly understood. During this presentation, we will first address the extent by which transcriptional signatures associated with MDD compares in men and women. We will then transition to the capacity of different mouse models of chronic stress to recapitulate some of the transcriptional alterations associated with the expression of MDD in both sexes. Finally, we will briefly elaborate on the functional consequences of these changes at the neuronal level and conclude with an integrative perspective on the contribution of sex-specific transcriptional profiles on the expression of stress responses and MDD in men and women.

SeminarNeuroscience

Vulnerable periods of brain development in ion channelopathies

Dirk Isbrandt
Deutsches Zentrum fur Neurodegenerative Erkrankunngen
Dec 15, 2020

Brain and neuronal network development depend on a complex sequence of events, which include neurogenesis, migration, differentiation, synaptogenesis, and synaptic pruning. Perturbations to any of these processes, for example associated with ion channel gene mutations (i.e., channelopathies), can underlie neurodevelopmental disorders such as neonatal and infantile epilepsies, strongly impair psychomotor development and cause persistent deficits in cognition, motor skills, or motor control. The therapeutic options available are very limited, and prophylactic therapies for patients at an increased risk of developing such epilepsies do not exist yet. By using genetic mouse models in which we controlled the activities of Kv7/M or HCN/h-channels during different developmental periods, we obtained offspring with distinct neurological phenotypes that could not simply be reversed by the re-introduction of the affected ion channel in juvenile or adult animals. The results indicate that channelopathy/mutation-specific treatments of neonatal and infantile epilepsies and their comorbidities need to be targeted to specific sensitive periods.

SeminarNeuroscienceRecording

Targeting the synapse in Alzheimer’s Disease

Johanna Jackson
UK Dementia Research Institute at Imperial College London
Dec 13, 2020

Alzheimer’s Disease is characterised by the accumulation of misfolded proteins, namely amyloid and tau, however it is synapse loss which leads to the cognitive impairments associated with the disease. Many studies have focussed on single time points to determine the effects of pathology on synapses however this does not inform on the plasticity of the synapses, that is how they behave in vivo as the pathology progresses. Here we used in vivo two-photon microscopy to assess the temporal dynamics of axonal boutons and dendritic spines in mouse models of tauopathy[1] (rTg4510) and amyloidopathy[2] (J20). This revealed that pre- and post-synaptic components are differentially affected in both AD models in response to pathology. In the Tg4510 model, differences in the stability and turnover of axonal boutons and dendritic spines immediately prior to neurite degeneration was revealed. Moreover, the dystrophic neurites could be partially rescued by transgene suppression. Understanding the imbalance in the response of pre- and post-synaptic components is crucial for drug discovery studies targeting the synapse in Alzheimer’s Disease. To investigate how sub-types of synapses are affected in human tissue, the Multi-‘omics Atlas Project, a UKDRI initiative to comprehensively map the pathology in human AD, will determine the synaptome changes using imaging and synaptic proteomics in human post mortem AD tissue. The use of multiple brain regions and multiple stages of disease will enable a pseudotemporal profile of pathology and the associated synapse alterations to be determined. These data will be compared to data from preclinical models to determine the functional implications of the human findings, to better inform preclinical drug discovery studies and to develop a therapeutic strategy to target synapses in Alzheimer’s Disease[3].

SeminarNeuroscienceRecording

Unravelling brain connectopathy in autism with cross-species fMRI

Alessandro Gozzi
Istituto Italiano di Tecnologia (Rovereto, Italy)
Nov 17, 2020
SeminarNeuroscience

The role of protein translation pathways in regulating excitation/inhibition balance in epilepsy

Carlo Sala
CNR (Milan, Italy)
Nov 3, 2020
ePoster

Disrupted Egocentric Vector Coding of Environmental Geometry in Alzheimer’s Disease Mouse Model

Yoonsoo Yeo, Jeehyun Kwag

COSYNE 2025

ePoster

VAME outperforms conventional assessment of behavioral changes and treatment efficacy in Alzheimer’s mouse models

Stephanie Miller, Kevin Luxem, Kelli Lauderdale, Pranav Nambiar, Patrick Honma, Katie Ly, Shreya Bangera, Nick Kaliss, Mary Bullock, Jia Shin, Yuechen Qiu, K Dakota Mallen, Zhaoqi Yan, Andrew Mendiola, Takashi Saito, Takaomi Saido, Alex Pico, Reuben Thomas, Erik Roberson, Katerina Akassoglou, Pavol Bauer, Stefan Remy, Jorge Palop

COSYNE 2025

ePoster

17β-estradiol and estrogen-like compound shows neuroprotective potential in a triple transgenic mouse model of Alzheimer’s disorder

Szidónia Farkas, Adrienn Szabó, Tamás Kovács, István M. Ábrahám, Dóra Zelena
ePoster

5-HT dynamics in a mouse model of depression

Kim Renken, Olivia Andrea Masseck

FENS Forum 2024

ePoster

Activation of NOTCH pathway in brain endothelial cells ameliorates vascular abnormalities in Alzheimer's disease mouse models

Silvia Quiñones Cañete, Juan Luis López Ogáyar, María Isabel Álvarez Vergara, David Macías, Alicia Elena Rosales Nieves, Alberto Pascual

FENS Forum 2024

ePoster

Adiponectin deficiency exacerbates cerebrovascular dysfunction in 5xFAD mouse model of Alzheimer’s disease

Wenying Zou, Leung-Wah Yick, Zifei Zhang, Jason Shing-Cheong Kwan, Koon-Ho Chan

FENS Forum 2024

ePoster

Adult cortical and hippocampal network dynamics in p.A263V Scn2a mouse model of developmental and epileptic encephalopathy

Yana Reva, Katharina Ulrich, Hanna Oelßner, Birgit Engeland, Ricardo Melo Neves, Stephan Marguet, Dirk Isbrandt

FENS Forum 2024

ePoster

Alfaxalone does not affect memory performance in a mouse model of Alzheimer’s disease

Nikola Milovanović, Irena Jovanović Macura, Vesna Tešić, Milka Perović, Vesna Pešić, Jelena Ćirić

FENS Forum 2024

ePoster

Alteration of myelin gene expression in a mouse model for Glut1 Deficiency Syndrome

Sophie Burlet-Godinot, Daphné Vorburger, Hubert Fiumelli, Maoxue Tang, Umrao Monani, Darryl De Vivo, Jean-Luc Martin, Pierre Magistretti

FENS Forum 2024

ePoster

Alterations in dopaminergic innervation and receptor expression in mouse models and patients with focal cortical dysplasia

Norisa Meli, Albert Becker, Sandra Blaess

FENS Forum 2024

ePoster

Altered circadian function in a mouse model of phenylketonuria

Junfei Cao, Sjoerd van Hasselt, Cecile Bruil, Wendy Hegge, Jorick te Velde, Eddy Van der Zee, Robbert Havekes, Peter Meerlo

FENS Forum 2024

ePoster

Altered excitatory/inhibitory balance in the prefrontal cortex of the IB2 KO mouse model of autism: From neuronal excitability to cerebellar modulation in vivo

Eleonora Pali, Danila Di Domenico, Maria Conforti, Ileana Montagna, Teresa Soda, Simona Tritto, Egidio D'Angelo, Francesca Prestori, Lisa Mapelli

FENS Forum 2024

ePoster

Altered hippocampal sharp-wave ripples play a role in impaired memory consolidation in Christianson syndrome mouse model

Jamie Mustian, Dhruv Mehrotra, Adrien Peyrache, Anne McKinney

FENS Forum 2024

ePoster

Analysis of brain network changes in a valproic acid-treated autism mouse model during social stimulation

Róbert Kemecsei, Viktória Horváth, András Csillag, Gergely Zachar

FENS Forum 2024

ePoster

Anterior cingulate cortex hyperexcitability in a mouse model of attention-deficit/hyperactivity disorder and pain comorbidity

Sandra Sanchez-Sarasua, Sarah Bou Sader Nehme, Marie Tuifua, Otmane Bouchatta, Marc Landry

FENS Forum 2024

ePoster

Antidepressant-like effects of psychedelics in a chronic despair mouse model: Is the 5-HT2A receptor the unique player?

Mehdi Sekssaoui, Joël Bockaert, Philippe Marin, Carine Bécamel

FENS Forum 2024

ePoster

Antipruritic effect of endogenously increased GABA by blockade of GAT1 in a mouse model of chronic itch

Mitsuo Tanabe, Misa Oyama, Yuuna Mase, Shun Watanabe, Takashi Iwai

FENS Forum 2024

ePoster

Application of dehydroepiandrosterone as a neuroprotective agent for the therapy of Alzheimer’s disease in a mouse model

Kvak Erika Eliza, Szidónia Farkas, Adrienn Szabó, Dóra Zelena

FENS Forum 2024

ePoster

Assessing the therapeutic potential of antidepressant and anti-inflammatory drugs in an inflamed depression mouse model: A comparative study of efficacy

Aurelia Viglione, Naomi Ciano Albanese, Giulia Fiorentini, Silvia Poggini, Anna Poleggi, Igor Branchi

FENS Forum 2024

ePoster

Assessment of Purkinje neuron degeneration in the flocculus vs. medial cerebellum in a mouse model of spinocerebellar ataxia type 13 (SCA13)

Anna Lena Langen, Oskar Markkula, Thanh Le, Rashmitha Senthilvel Selvakumar, Ian D. Forsythe, Conny Kopp-Scheinpflug

FENS Forum 2024

ePoster

An atopic dermatitis mouse model reveals potential utility for atopic dermatitis-generated comorbid depression brain circuitry studies

Ian McConnell, Bhuvana Chimmiri, Santosh Mishra

FENS Forum 2024

ePoster

Gut bacterial toxin further enhances blood-brain barrier alterations in a progressive mouse model of Parkinson’s disease

Kristina Lau, Lisa Porschen, Anna-Sophia Hartke, Christopher Käufer, Birthe Gericke, Franziska Richter

FENS Forum 2024

ePoster

Behavioral and histological hallmarks of an intrastriatal rotenone mouse model for Parkinson’s disease

Paula Jauregi Barandica, Roland Rabl, Magdalena Daurer, Livia Breznik, Stefanie Flunkert, Boris Philippe Chagnaud, Manuela Prokesch

FENS Forum 2024

ePoster

Behavioral, molecular and cellular effects of low-dose CBD administration in a chronic stress-induced major depression mouse model

Sara Borràs Pernas, Anna Sancho-Balsells, Daniel Del Toro, Albert Giralt

FENS Forum 2024

ePoster

A behavioural assessment to characterize different stages of memory impairment in humanized APP knock-in mouse models across various ages

Loukia Katsouri, Angela Misak, Stephen Burton, Jade Sangha, John O'Keefe

FENS Forum 2024

ePoster

Blood-spinal cord barrier alterations in a mouse model of centrifugation-induced hypergravity

David Dubayle, Nicolas Rebergue, Yann Godfrin, Jean-Luc Morel, Sighild Lemarchant

FENS Forum 2024

ePoster

Brain activation during inflammatory and non-inflammatory hyperalgesia in a rheumatoid arthritis mouse model

Rita Gálosi, Gábor Kriszta, Éva Borbély, Ádám I Horváth, Valéria Tékus, Nikolett Szentes, Zsuzsanna Helyes

FENS Forum 2024

ePoster

Brain delivery and efficacy of a BBB shuttle-enhanced aducanumab biosimilar in a mouse model of Alzheimer’s disease

Thomas Topilko, Marta Ramos Vega, Franziska Wichern, Allan Jensen, Camilla Stampe Jensen, Jacob Lercke Skytte, Casper Graversen Salinas, Sandra Vergo, Jacob Hecksher-Sørensen, Henrik H. Hansen

FENS Forum 2024

ePoster

Brain-state dependent deficit in visual discrimination in a mouse model of SYNGAP1-related intellectual disability and autism

Danai Katsanevaki, Nathalie Dupuy, Sam Booker, Nina Kudryashova, Damien Wright, Aisling Kenny, Zihao Chen, Pippa Howitt, Steffen Schneider, Mackenzie Mathis, Andrew Stanfield, Peter Kind, Nathalie Rochefort

FENS Forum 2024

ePoster

Cellular and molecular characterization of serotonergic synapses in a mouse model of depression and raphe synucleinopathy

Unai Sarriés-Serrano, Lluis Miquel-Rio, Sarka Jelinkova, Vincent Paget-Blanc, Verónica Paz, J Javier Meana, Etienne Herzog, Analia Bortolozzi

FENS Forum 2024

ePoster

Cerebellar alteration in a mouse model of GRIN2D-related developmental and epileptic encephalopathies

Mor Yam, Danielle Galber, Wayne N. Frankel, Karen B. Avraham, Moran Rubinstein

FENS Forum 2024

ePoster

Cerebellar BDNF signaling downregulation and autistic-like traits: Insights from a cholesterol storage disorder mouse model

Greta Massa, Serena Camuso, Lucy Babicola, Roberta Stefanelli, Jessica Tiberi, Piergiorgio La Rosa, Maria Teresa Fiorenza, Sonia Canterini

FENS Forum 2024

ePoster

Changes in endocannabinoid-dependent synaptic plasticity in CA1 hippocampus of a mouse model of temporal lobe epilepsy

Amaia Mimenza, Itziar Bonilla-Del Río, Izaskun Elezgarai, Nagore Puente, Pedro Grandes

FENS Forum 2024

ePoster

Changes in striatal spiny projection neurons’ properties and circuitry in a mouse model of autism spectrum disorder with cholinergic interneuron dysfunction

Juliette Graniou, Xavier Caubit, Pascal Salin, Lydia Kerkerian-Le Goff, Laurent Fasano, Paolo Gubellini

FENS Forum 2024

ePoster

Characterization of early post-natal development and ultrasonic vocalizations in mouse models of GRIN1 disorder

Megan Sullivan, Wendy Horsfall, Ali Salahpour, Amy J. Ramsey

FENS Forum 2024

ePoster

Characterization of the expression of dopaminergic markers in the Cntnap2 knockout mouse model of autism

María Prieto, Olga Peñagarikano

FENS Forum 2024

ePoster

Characterization of a novel mouse model for CHD2-related neurodevelopmental disorder

Anat Mavashov Arzuan, Shaked Turk, Marina Brusel, Shir Quinn, Yael Sarusi, Igor Ulitsky, Moran Rubinstein

FENS Forum 2024

ePoster

Characterization of the pathophysiological mechanisms of KCNQ2-developmental and epileptic encephalopathy (KCNQ2-DEE) in the KV7.2Thr274Met/+ mouse model

Shaimaa Haiba, Kilian Lüdicke, Laurent Villard, Maurizio Taglialatela, Carmine Ostacolo, Holger Lerche, Thomas V. Wuttke

FENS Forum 2024

ePoster

Characterization of the transcriptional landscape of endogenous retroviruses at the fetal-maternal interface in a mouse model of autism spectrum disorder

Martina Giudice, Antonella Camaioni, Anna Maria Tartaglione, Vita Petrone, Claudia Matteucci, Gemma Calamandrei, Paola Sinibaldi-Vallebona, Laura Ricceri, Emanuela Balestrieri, Chiara Cipriani

FENS Forum 2024

ePoster

Exploring the neuroprotective effect of auditory enhanced slow-wave sleep in a mouse model of Alzheimer’s disease

Inês Dias, Irena Barbaric, Vera Gysin, Christian Baumann, Sedef Kollarik, Daniela Noain

FENS Forum 2024