DNM 24 DNM 24

Dutch
Neuroscience Meeting

Dutch
Neuroscience Meeting

Tiel, The Netherlands • 13-14 June 2024

Tiel, The Netherlands
13-14 June 2024

Preliminary Programme

  • Thursday, 13 June
  • Friday, 14 June
  • 09:00 Registration (with Coffee & Tea)

    10:00 Opening

    10:10 Plenary Session
    Session 1: Keynote Lecture
    Chair: Harold MacGillavry (Utrecht)

    Rick Huganir  (Baltimore, MD, USA) Regulation of neurotransmitter receptors during synaptic plasticity in health and disease (45')

    11:00 Coffee & Tea

    11:30 Parallel Sessions A
    Session 2: Mitochondria at the crossroads between energy hub and signaling in health and disease
    Chairs: Amalia Dolga (Groningen) & Nael Nadif Kasri (Nijmegen)

    György Hajnóczky  (Philadelphia, PA, USA) Mitochondrial calcium dysregulation and neurodegeneration (30')
    Vivi Heine  (Amsterdam) Mitochondrial changes in schizophrenia patient iPSC-derived astrocytes (15')
    Christian Lohmann  (Amsterdam) Mitochondrial dynamics during synapse development (15')
    Werner Koopman  (Nijmegen) Whole-body Ndufs4 knockout mice with isolated complex I deficiency engage a futile adaptive brain response (15')
    Amalia Dolga  (Groningen) Targeting mitochondrial calcium uniporter complex in human microglia and brain organoids derived from iPSC of patients with Alzheimer's disease   (15')

    Session 3: Progress in translational approaches for experimental aggression research
    Chairs: Rudy Schreiber & Jill Lobbestael (Maastricht)

    Jozsef Haller  (Budapest, Hungary) Neurobiology of aggression (30')
    Jill Lobbestael  (Maastricht) The psychology of aggression (15')
    Jocelien Olivier  (Groningen) Swift release, fierce nature? The link between ejaculation speed and aggression in wildtype Groningen rats (15')
    Fedde Sappelli  (Nijmegen) Provocation and assessment of aggression using VR (15')
    Camiel van der Laan  (Amsterdam) A genetically informed investigation of aggression subtypes (15')

    Session 4: Early changes in myelin structure and lipid composition in multiple sclerosis: toward a new era beyond the classic autoimmunity hypothesis?
    Chairs: Antonio Luchicchi & Gijs Kooij (Amsterdam)

    Markus Kipp  (Rostock, Germany) The sphingosin-1-phosphate receptor signaling cascades: Common pathways for immunosuppression and oligodendrocyte protection (30')
    Martin Giera  (Leiden) A lipidomics view on neurodegeneration (15')
    Serhii Chornyi  (Amsterdam) Redirection of cholesterol biosynthesis pathway as a potential MS therapy (15')
    Gema Munoz-Gonzalez  (Amsterdam) Human post-mortem organotypic brain slice cultures: a novel tool to study intrinsic mechanisms of MS pathogenesis (15')
    Aletta van den Bosch  (Amsterdam) Ultrastructural axon-myelin unit alterations in MS and implications for MS lesion formation (15')

    Session 5: TMS in psychiatry
    Chairs: Odile van den Heuvel (Amsterdam) & Chris Baeken (Ghent, Belgium)

    Chris Baeken  (Ghent, Belgium) Neurobiological aspects of accelerated rTMS in depression (30')
    Sophie Fitzsimmons  (Amsterdam) Multilayer brain network properties of the stimulation location predict outcome of repetitive transcranial magnetic stimulation for obsessive-compulsive disorder (15')
    Eva Dijkstra  (Amsterdam) TMS-induced heart-brain coupling: implications for TMS target stratification (15')
    Debby Klooster  (Eindhoven) MRI-based targeting rTMS (15')
    Iris Dalhuisen  (Nijmegen) rTMS as a treatment for depression: current practice and new developments (15')

    Session 6: Sex hormones and the circadian clock
    Chairs: Dirk Jan Stenvers & Joram Mul (Amsterdam)

    Jennifer Evans  (Milwaukee, WI, USA) Sex differences in circadian clock function and responses to light (30')
    Margot Morssinkhof  (Amsterdam) Chronotype changes after sex hormone use: A prospective cohort study in transgender users of gender-affirming hormones (15')
    Raymond Noordam  (Leiden) Estradiol levels and circadian sleep-wake behaviour: data from the UK Biobank (15')
    Dirk Jan Stenvers  (Amsterdam) Effect of estradiol levels on sleep-wake rhythms in the Netherlands Epidemiology of Obesity study (15')
    Ayano Shiba  (Amsterdam) Exercise and the brain clock; effects of voluntary running on SCN ∆FosB in male and female Wistar rats (15')

    Session 7: From birth to disease: cell-extrinsic microenvironmental regulation of brain cell fate
    Chairs: Marloes Verkerke (Amsterdam) & Emma van Bodegraven (Utrecht)

    Fiona Doetsch  (Basel, Switzerland) Regulation and diversity of adult neural stem cells (30')
    Carlos Flores Clara  (Utrecht) Cell cities: unusual crosstalk between Interneurons and oligodendrocyte precursor cells in brain development (15')
    Nils Bessler  (Utrecht) Pontine regionalized brain organoids model human diffuse midline glioma (15')
    Lucas Baudouin  (Amsterdam) Using patient-specific multiscale imaging to elucidate the role of oligodendroglial abnormalities in cognitive impairment in glioma. (15')
    Loet Coenen  (Amsterdam) Periphery-Brain interaction in Aging and Dementia – A bloody interesting story (15')

    13:00 Lunch

    14:15 Parallel Sessions B
    Session 8: Advances in neurodevelopmental ressearch and associated disorders
    Chairs: Catarina Osorio & Martijn Schonewille (Rotterdam)

    Lilian Kisiswa  (Aarhus, Denmark) The role of mutant O-GlcNAc transferase in synaptic dysfunction and intellectual disability (30')
    Heiling Lu  (Rotterdam) Functional significance of differentially expressed genes in subpopulations of Purkinje cells (15')
    Jeroen Pasterkamp  (Utrecht) Novel molecular mechanisms in neural circuit development (15')
    Hilde Smeenk  (Rotterdam) Increased GABAergic neurogenesis in human cortical organoids with schizophrenia-associated SETD1A mutations (15')
    Nael Nadif Kasri  (Nijmegen) Leveraging spontaneous activity in human stem cell derived neurons to model neurodevelopmental disorders (15')

    Session 9: Pollutants2.0: Unraveling the sensing and signaling power of environmental stressors
    Chairs: Martina Schmidt (Groningen) & Jacco Briedé (Maastricht)

    Lucia Migliore  (Pisa, Italy) Gene-environment interactions in Alzheimer disease: the emerging role of epigenetics (30')
    Jacco Briedé  (Maastricht) Transcriptomics approaches used to investigate the molecular mechanisms that form the link between environmental pollutants and Alzheimer’s disease (15')
    Martine Smit  (Amsterdam) Viral G protein-coupled receptors modulate glioblastoma via extracellular vesicles (15')
    Karim Rafie  (Groningen) Imaging pollution - Using cryo-EM to elucidate the events affecting the brain by diesel exhaust particles. (15')
    Martina Schmidt  (Groningen) Unravelling the sensing and signaling power of pollutants (15')

    Session 10: Genetics and multi-omics to dissect causes of psychiatric disease
    Chairs: Sabine Spijker (Amsterdam) & Inge Holtman (Groningen)

    Guus Smit  (Amsterdam) Proteomics analysis of frontal cortex of schizophrenia patients (30')
    Constanze Seidenbecher  (Magdeburg, Germany) Cerebrospinal fluid levels of neural extracellular matrix components in schizophrenia (15')
    Femke Roig-Kuhn  (Amsterdam) Proteomics to dissect the molecular mechanisms underlying major depressive and bipolar disorder (15')
    Tommaso Fortunato Asquini  (Groningen) Integrating transcriptomic and phenotype data to dissect the molecular mechanisms underlying common psychiatric disorders (15')
    Kitty Reemst  (Tel Hai, Israel) Early-life stress lastingly impacts microglial transcriptome and function under basal and immune-challenged conditions (15')

    Session 11: SUPERGLUE: how SUbcellular comPartments in astroGLia control cognitive processing
    Chairs: Mark Verheijen & Rogier Min (Amsterdam)

    Christian Henneberger  (Bonn, Germany) Adaptive and maladaptive rapid remodelling of perisynaptic astroglia (30')
    Aline Mak  (Amsterdam) Astro-eGRASP: assessing astrocyte-synapse interaction on persistent memory engram cells (15')
    Quinty Bisseling  (Amsterdam) Fragility of cytoskeleton-membrane interactions in astrocytes lacking MLC1; implications for the rare leukodystrophy MLC (15')
    Stephanie Beekhuis-Hoekstra  (Amsterdam) Modelling the interaction of brain microvascular endothelial cells and astrocytes in vitro (15')
    Nina Smets  (Amsterdam) The perivascular space: in between the astrocyte and the blood vessel (15')

    Session 12: Enhancing efficacy in cross-cultural collaboration (sponsored by World Women in Neuroscience & IBRO)
    Chairs: Jocelien Olivier (Groningen) & Wanne Wiersinga (Amsterdam)

    Session 13: Individual differences in drug addiction: from mechanisms to treatment
    Chairs: Cyprien Guerrin & Arnt Schellekens (Nijmegen)

    Bart Ellenbroek  (Wellington, New Zealand) The transgenerational effects of alcohol (30')
    Nathan Marchant  (Amsterdam) The role of the anterior insula cortex in choice between alcohol and social reward (15')
    Rick Hesen  (Nijmegen) Increased cocaine self-administration and prefrontal cortical dysregulation of glutamatergic and GABAergic signalling in a rat model for sensory processing sensitivity (15')
    Debbie Tesselaar  (Nijmegen) Neurobiological markers of substance use disorder and co-occurring non-addiction psychiatric comorbidities: a systematic review (15')
    Emese Kroon  (Rotterdam) Cross cultural neuroscience: the role of perceived harms and benefits of use in cannabis use disorder (15')

    15:45 Coffee & Tea

    16:00 Plenary Session
    Session 14: Keynote Lecture
    Chair: Martijn Schonewille (Rotterdam)

    Mathias  Jucker  (Tübingen, Germany) Proteopathic seeds in neurodegenerative diseases (45')

    17:00 Posters & drinks
    Session 15: Poster session 1

    18:30 Dinner

    20:00 Plenary Session
    Session 16: Brain Bee, Art of Science, Top paper, Thesis & Young Talent Prizes

    21:00 Social

    Keynote Lecture

    Mitochondria at the crossroads between energy hub and signaling in health and disease

    Mitochondria are central to all essential cellular processes governing the balance between death and survival. Our session, featuring leading experts in mitochondrial biology, explores the pivotal role of mitochondrial function in brain health and disease. This session will spotlight the interplay between mitochondrial calcium flux and cellular bioenergetics, dissecting its implications for neuronal survival and pathology. The speakers will show how mitochondrial dynamics at the synapse can influence synaptic strength and plasticity. Utilizing patient-derived iPSCs, speakers will demonstrate how these cellular models are revolutionizing our understanding on cell metabolism in models of neurodegenerative processes. Cutting-edge high-resolution imaging techniques and specialized sensors will reveal the elusive mito-ER contact sites, providing insights into mitochondrial communication hubs that dictate cell survival and function. Our session aims to foster a cross-disciplinary dialogue, bridging gaps between molecular mechanisms and translational applications, and to inspire innovative therapeutic strategies targeting mitochondrial dysfunction in brain disorders.

    Progress in translational approaches for experimental aggression research

    Aggression is a major issue in society that receives only marginal attention in research. People working in clinics, care homes and forensic settings are exposed to aggressive behaviours on a daily basis. Recent numbers in the Netherlands indicate that about 75% of the health and social work workers were confronted with aggressive behaviours. There is a need to better characterize the genetic, neurobiological and psychological underpinnings of aggressive behaviours in order to develop improved measures for prevention and containment. This symposium will breach this topic from a translational neuroscience perspective, covering vertical, that is, between rodents and humans; as well as, horizontal translation, that is, between healthy and impaired. The main aim is to provide an overview of recent developments on: 1) neural mechanisms of aggression and violence in animal models with a focus on the serotonergic system; 2) the use of virtual reality in human experimental behavioural sciences to study reactive aggression; and 3) genetic studies in aggression. Bringing researchers from these different fields together may provide inspiration for new studies and, ideally, novel ideas for the prevention and treatment of aggressive behaviours.

    Early changes in myelin structure and lipid composition in multiple sclerosis: toward a new era beyond the classic autoimmunity hypothesis?

    The cause of multiple sclerosis (MS) is still unknown. However, in recent years, studies are emerging that challenge the classic vision of MS as a pure immune cell driven disease. In turn, an increasing number of scientists are posing an alternative hypothesis, indicating that early changes at the level of normal appearing white matter (NAWM) myelin proteins and lipids may act as a primary event to instigate the cascade of pathological events leading to the formation of overt focal MS lesions within the central nervous system. In this interdisciplinary session with speakers from different institutes and at different stages of their career (who have all confirmed their interest), we will discuss how the latest findings related to these subtle changes at the level of NAWM myelin may play a role in MS pathogenesis ranging from myelin lipid biochemistry to ultrastructural myelin morphology and in vivo findings. Ultimately, we aim to pave the way to apply such alterations as novel biomarkers as well as potential targets for intervention in the context of MS.

    TMS in psychiatry

    Sex hormones and the circadian clock

    Women are earlier chronotypes then men, and this difference disappears after menopause, when estradiol production stops. Early chronotypes have lower risks of obesity, dyslipideamia and type 2 diabetes mellitus than late chronotypes, so this may affect postmenopausal health. A potential explanation of the sex difference in chronotypes is an effect of the sex hormone estradiol on the central brain clock in the suprachiasmatic nucleus, as this brain region has high expression of the main estrogen receptor. In this session, an excellent team of researchers will present their latest findings how sex differences and estradiol impact the central brain clock in fundamental animal studies, in the transgender population, and on an epidemiological level.

    From birth to disease: cell-extrinsic microenvironmental regulation of brain cell fate

    Whether a cell proliferates, differentiates, migrates or dies is dependent on cell-intrinsic and -extrinsic factors. Regulating these various cell fates are key components to brain development and deregulation leads to disease. It is becoming increasingly recognized that spatially and temporally defined microenvironments within the brain largely impact cell fate. Our first speaker, Carlos Flores Clara, investigates the cellular interactions of migrating neurons with their surroundings during development. In the adult brain, neuro- and gliogenesis is dramatically decreased. Our keynote speaker Prof. Dr. Fiona Doetsch discovered specialized gliogenic domains in the ventral subventricular zone of the adult mouse brain which become more active in response to demyelination. Deregulation of neuro- and gliogenesis can initiate brain tumor growth and is largely impacted by the microenvironment. Dr. Anoek Zomer discovered treatment-induced microenvironmental changes which impact brain tumor growth. Dr. Lucas Baudouin will discuss how brain tumor-induced changes in the microenvironment affect neuron-glia interactions. Finally, Loet Coenen will present on blood-derived factors that impact the brain’s microenvironment and neurodegeneration.

    Advances in neurodevelopmental ressearch and associated disorders

    Human brain development has a protracted time course that starts during gestation and lasts until early adulthood. During this extended period, several processes such as neurogenesis, migration, axon guidance, differentiation and synaptogenesis need to be tightly regulated in time and space. Small disruptions in any of these processes can lead to devastating neurological disorders such as intellectual disability, attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorder, and schizophrenia. Therefore, understanding early molecular and cellular mechanisms regulating the nervous system is imperative to comprehend brain disorders and provide new avenues for novel therapeutic interventions. The goal of this parallel session is to highlight the latest findings on key molecular and cellular processes involved in nervous system development, the pathways affected when development is disrupted, and the latest methodologies to study these questions. To this end we have invited experts in the neurodevelopmental field at different stages of their career. Given the broad range of backgrounds, brain regions and career stages of the speakers, we are confident that this parallel session will appeal to a broad audience.

    Pollutants2.0: Unraveling the sensing and signaling power of environmental stressors

    Exposure to environmental pollutants contributes to diverse disease pathology including cancer, stroke, Alzheimer’s disease and dementia. Pollutants entry can occur through inhalation, traversing endothelial and epithelial barriers, and crossing the blood-brain barrier, leading to a wide distribution throughout the human body via systemic circulation. Pollutants cause cellular damage by overarching mechanisms encompassing oxidative stress, mitochondrial dysfunction, (neuro)inflammation, and protein instability. Sensing Pollutant has added a new dimension to disease progression and drug failure. Understanding their molecular pathways and potential receptor binding or signaling could contribute to ways to combat their detrimental effects. We highlight key points of pollutants signaling, and crosstalk to receptors acting as drug targets for chronic diseases, and the potential for future therapeutic avenues. The selected speakers will provide novel insights into underlying mechanisms. As such our session aims to create a very timely platform for the cross-fertilization of diverse disciplines and (technical) approaches using (epi)genetics, (proteo)omics, x-ray crystallography and cryogenic electron microscopy.

    Genetics and multi-omics to dissect causes of psychiatric disease

    Mood disorders represent a major burden on public health, ranking third in the leading causes of disability worldwide. However, research efforts have not discovered quantitative measures for its diagnosis, nor pharmaceutical interventions that are widely effective. This makes studying mood disorders a difficult and interesting problem that requires innovative and interdisciplinary solutions. Molecular understanding of the disease through genetics and genomics, in combination with deep-phenotyping (phenomics) of both (pre-)clinical features and environmental factors, is promising. Therefore, we focus in this session on recent efforts to dissect underlying molecular mechanisms and ways to predict clinical outcomes in human subjects and animal models of depression. As a main speaker, we propose Dr. Na Cai, known for her work on dissecting phenotype-genotype relations (Nature Genetics 2020, 2023). We invite her to talk about her latest collaborative Nature Genetics paper to improve GWAS power focusing on generating biological insight by making better use of phenonomics data rather than only increasing sample size. The 4 other speakers will present various -omics aspects in (models of) depression using different species.

    SUPERGLUE: how SUbcellular comPartments in astroGLia control cognitive processing

    The crucial role of glial cells, in particular astrocytes, in higher brain functions is increasingly recognized. Astrocytes have a complex morphology, forming two types of specialized subcellular compartments: astrocyte endfeet enwrapping the brain vasculature and perisynaptic astrocyte processes (PAPs) surrounding neuronal synaptic contact points. This places astrocytes in a unique position, potentially enabling them to bridge regulation of brain fluid homeostasis to control over neuronal signal transmission. But insight into the establishment and dynamic regulation of this astrocyte bridge function, and how it is involved in cognitive processing is lacking. The main aim of this symposium is therefore to bring together experts on astrocyte-vasculature interactions with experts on astrocyte-synapse interactions. This combination will enable to discus, together with the audience, the complex bridge function of astrocytes and how they orchestrate specialized subcellular compartments to optimize brain information processing. This proposal is timely, since the proposed speakers each have recently developed unique state-of-the-art techniques, instruments and model systems to break this new ground.

    Enhancing efficacy in cross-cultural collaboration (sponsored by World Women in Neuroscience & IBRO)

    Individual differences in drug addiction: from mechanisms to treatment

    Addiction affects millions of people worldwide and is a leading cause of disability. Current treatments are moderately effective and fail in addressing this major problem partly due to the large clinical heterogeneity. To move forward, we must shift from generic to personalized treatments, tailoring interventions to individual needs. This requires a deep understanding of the behavioral and neurobiological mechanisms underlying this heterogeneity. This session explores clinical and preclinical strategies aiming to understand individual differences in drug addiction, addressing both behavioral and neurobiological levels. This session covers diverse approaches, from examining cross-cultural attitudes in brain processes through fMRI (approach-avoidance, cue-reactivity, cognitive control), to measuring neurotransmitter systems (dopaminergic and serotoninergic systems) using PET imaging and recording neuronal activity with fiber photometry. This session also investigates comorbidities, transgenerational alcohol effects, and choice preference between alcohol and social reward. Commonalities across model systems as well as the pathophysiology of addiction development for predicting and developing preventive approaches will be discussed.

    Keynote Lecture

    Poster session 1

    Brain Bee, Art of Science, Top paper, Thesis & Young Talent Prizes

  • 09:00 Registration (with Coffee & Tea)

    09:30 Parallel Sessions C
    Session 17: Molecular mechanisms in neurodegeneration
    Chairs: Hannah Walgrave & Marijn Kuijpers (Nijmegen)

    Bart De Strooper  (Leuven, Belgium) The cellular phase of Alzheimer’s Disease and the search for new drug targets.
      (30')

    Erik Storkebaum  (Nijmegen) Peripheral neuropathy caused by mutant tRNA synthetases: from molecular mechanism to gene therapy
      (15')

    Amber Penning  (Amsterdam) Systematic assessment of microRNA-132 therapeutic targeting in Alzheimer’s disease using iPSC-derived microglia (15')
    Elly Hol  (Utrecht) Alexander disease and the astrocyte intermediate filament protein GFAP (15')
    Eline van Hugte  (Nijmegen) Early alpha synuclein dysfunctions in a human model for Parkinson’s disease (15')

    Session 18: Unlocking the gut-brain connection in aging: from in vitro studies to clinical implications and the role of dietary interventions
    Chairs: Joukje Oosterman (Nijmegen) & Mara van Trijp (Wageningen)

    Esther Aarts  (Nijmegen) The importance of the gut-brain axis for neurocognition (30')
    Aina Casademont-Roca  (Wageningen) The protective effects of berries on metabolic endotoxemia in an in vitro gut-immune cell model (15')
    Lianne Remie  (Nijmegen) The role of microbial-derived short-chain fatty acids in neurocognitive ageing (15')
    Yannick Vermeiren  (Wageningen) Exploring prebiotics for cognitive resilience: state of the evidence and future directions (15')
    Kirsten Krüger  (Wageningen) The effect of the Mediterranean diet on the gut-brain axis in older adults: analysing host-gut microbiota co-metabolism of tryptophan. (15')

    Session 19: Computational genomics approaches to study disease mechanisms
    Chairs: Inge Holtman (Groningen) & Onur Basak (Utrecht)

    Alexi Nott  (London, United Kingdom) Epigenetic mechanisms underlying age-related brain disorders (30')
    Ahmed Mahfouz  (Leiden) Decoding cell-type-specific exon inclusion in the human brain (15')
    Lotje de Witte  (Nijmegen) The human microglia responsome: a resource to better understand microglia states in health and disease (15')
    Charlotte van Dijk  (Utrecht) Exploring Single-Cell Alternative PolyAdenylation in Neurodegenerative Disorders (15')
    Laleh Haghverdi  (Berlin, Germany) Temporal dynamics in single-cell omics data & comparative multi-patient multi-modal data visualisation  (15')

    Session 20: Microglial functional and neuroanatomical heterogeneity in diseases
    Chairs: Chun-Xia Yi & Felipe Correa da Silva (Amsterdam)

    Tuan Leng Tay  (Boston, MA, USA) Microglial fate in neurodegeneration: for better or worse? (30')
    Shanshan Guo  (Shenzhen, China) Reprogram microglial function in the brain through the delivery of siRNA and compounds using nanoparticle technology (15')
    Felipe Correa da Silva  (Amsterdam) Microglial phagolysosome dysfunction and altered neural communication in Prader-Willi Syndrome (15')
    Han Jiao  (Amsterdam) Unresolved: Time-Restricted Feeding's Limitations on Microglial Function in Diet-Induced Obesity (15')
    Sara Pecoraro  (Amsterdam) Subventricular zone microglia promote a protective niche for neuronal progenitors in Parkinson’s disease (15')

    Session 21: Multi-regional perspective of sensorimotor integration in health and disease
    Chairs: Zhenyu Gao & Xiaolu Wang (Rotterdam)

    Ian Duguid  (Edinburgh, United Kingdom) Neural circuits for executing task-specific movements (30')
    Guy Bouvier  (Saclay, France) Inter- and Intra-hemispheric Sources of Vestibular Signals to V1 (15')
    Lizz Fellinger  (Amsterdam) Wave-like striatal dopamine peaks and troughs bracket spontaneous movement (15')
    Xiaolu Wang  (Rotterdam) Projection motifs of cerebellar modules underlying sensorimotor behaviors (15')
    Djaina Satoer  (Rotterdam) Awake brain surgery and diagnosis of aphasia in brain tumor patients (15')

    Session 22: Human cell modelling of human brain development and associated disorders
    Chairs: Adrian Harwood (Maastricht) & Vivi Heine (Amsterdam)

    Giuseppe Testa  (Milan , Italy) Human neurodiversity in a dish: towards a paradigm shift for neurodevelopmental modelling through organoid epidemiology (30')
    Jennifer Ramautar  (Amsterdam) The effects of L-serine in patients with GRIN2B related neurodevelopmental disorders: an EEG study (15')
    Femke de Vrij  (Rotterdam) Using human induced pluripotent stem cell derived neural models to test antisense oligonucleotide therapeutic applications (15')
    Bart Rutten  (Maastricht) Chronic cortisol induces alterations in neuronal processes at different stages of neuronal development in human embryonic stem cell-derived cortical neurons (15')
    Anna Wiersema  (Utrecht) ALS-associated mutations in FUS cause axonal changes in iPSC-derived spinal motor neurons (15')

    11:00 Coffee & Tea

    11:15 Parallel Sessions D
    Session 23: Neurological biomarkers of cancer-related cognitive impairment and fatigue: connecting cancer, (neuro)inflammation and behavior
    Chairs: Marieke van der Schaaf & Charlotte Sleurs (Tilburg)

    Sabine Deprez  (Leuven, Belgium) Insights from advanced neuroimaging on cancer-related cognitive impairment after breast cancer treatment (30')
    Rudi D’Hooge  (Leuven, Belgium) Preclinical models and experimental therapeutics in chemotherapy-induced cognitive impairment (15')
    Laura Kuil  (Amsterdam) The role of glia and cellular senescence in radiotherapy-induced cognitive impairment (15')
    Eline van Roekel  (Maastricht) Longitudinal associations of metabolites of the tryptophan-kynurenine pathway and other metabolites with fatigue severity up to two years after colorectal cancer treatment (15')
    Marieke van der Schaaf  (Tilburg) The effects of acute systemic inflammation on fatigue and cost-benefit decision making and its relevance for cancer-related fatigue (15')

    Session 24: Modulation of the axon initial segment as a remote control of neuronal activity
    Chairs: Amelie Freal & Viktor Al Naqib (Amsterdam)

    Maren Engelhardt  (Linz, Austria) Revisiting axonal plasticity from the nanoscale up (30')
    Amelie Freal  (Amsterdam) Local control of membrane trafficking drives axon initial segment plasticity (15')
    Viktor Al Naqib  (Amsterdam) The role of activity-dependent microglia-AIS interactions (15')
    Ana Rita Cunha  (Amsterdam) Uncovering the neuron type specificity of AIS plasticity mechanisms (15')
    Lia Carvalhais  (Nijmegen) Unraveling in the roles of the cisternal organelles in the axon initial segment (15')

    Session 25: Neurobiological substrates of sex differences in anxiety and stress
    Chairs: Sabrina van Heukelum (Nijmegen) & Jocelien Olivier (Groningen)

    Katharina Gryksa  (Regensburg, Germany) Social fear conditioning in rats and mice: Differences in social trauma susceptibility between sex and strains (30')
    Janine Doorduin  (Groningen) Sex differences in the behavioural and brain response to maternal immune activation (15')
    Michael Vencer Malaluan  (Amsterdam) Sex differences in the hypothalamic gene expression in response to stress exposure in early life and in adulthood (15')
    Jocelien Olivier  (Groningen) Stress prior to pregnancy and perinatal fluoxetine treatment exert sex-specific effects on the affective and social behaviour of rat offspring (15')
    Marloes Henckens  (Nijmegen) Whole-brain neuronal activity supporting the response to acute stress; a matter of sex?    (15')

    Session 26: Wrapping the brain: oligodendrogenesis and myelination in health and disease
    Chairs: Simone Mesman & Vivi Heine (Amsterdam)

    Melanie Küspert  (Erlangen, Germany) Timing is everything: Downstream effectors of Sox10 and their role as timers of oligodendroglial differentiation and CNS myelination (30')
    Liza Kok  (Amsterdam) Studying neuron-glia interactions in hypomyelinating disorder 4H leukodystrophy using iPSC- derived models (15')
    Simone Mesman  (Amsterdam) Tcf4 in regulation of oligodendrogenesis and myelination of the murine brain (15')
    Wendy Oost  (Groningen) Uncovering excitatory synapses in white matter multiple sclerosis lesions: a sign of neurogenesis (15')
    Rosanne Govaarts  (Leiden) Probing diffusion of water and metabolites to assess white matter microstructure in Duchenne muscular dystrophy (15')

    Session 27: Bidirectional insights: how neuroscience and machine learning inform and transform each other
    Chairs: Abdel Rayan & Yuliya Shapovalova (Nijmegen)

    Sander Bohte  (Amsterdam) Attention-gated brain propagation: how the brain can implement reward-based error backpropagation (30')
    Jorge Mejias  (Amsterdam) Diverse and flexible behavioral strategies in recurrent neural networks trained on decision making tasks (15')
    Parisa Naseri  (Nijmegen) Integrative machine learning framework for predicting cognitive decline from task-based fMRI data (15')
    Abdel Rayan  (Nijmegen) Advancing rodent sleep phase identification with deep neural networks and improved feature extraction methods (15')
    Devika Narain  (Rotterdam) Neural manifold discovery using low-distortion Riemannian alignment of tangent spaces (15')

    Session 28: Identifying novel neurobiological targets for the development of innovative treatment strategies for substance use disorders
    Chairs: Mieke Schulte (Rotterdam) & Anne Marije Kaag (Amsterdam)

    Ravi Das  (London, United Kingdom) Silencing maladaptive memories: a new translational approach to treating addiction? (30')
    Willeke van Dijk  (Amsterdam) Heart rate variability-biofeedback to support pregnant women quit smoking via stress reduction (15')
    Mieke Schulte  (Rotterdam) Deep TMS of the insula for addiction treatment (15')
    Kenan de Leeuw  (Amsterdam) Therapeutic use of psychedelics for substance use disorders (15')
    Anne Marije Kaag  (Amsterdam) Sex and gender-specific targets for the treatment of substance use disorders (15')

    12:45 Lunch

    14:00 Plenary Session
    Session 29: Keynote Lecture
    Chair: Marloes Henckens (Nijmegen)

    Isabelle Mansuy  (Zürich, Switzerland) Epigenetic inheritance: how traumatic experiences in early life can affect descendants via the germline (45')

    14:45 Coffee & Tea

    15:00 Parallel Sessions E
    Session 30: Interneuron dynamics during brain development and maturation in health and disease
    Chairs: Carla Gomes da Silva (Utrecht) & Corette Wierenga (Nijmegen)

    Juan Burrone  (London, United Kingdom) The emergence and plasticity of inhibitory synapses: from dendrites to the axon initial segment (30')
    Corette Wierenga  (Nijmegen) What is the role of the GABA shift in brain development? (15')
    Martijn Selten  (London, United Kingdom) Mechanisms of homeostatic regulation in parvalbumin-positive interneurons (15')
    Moritz Negwer  (Nijmegen) Brain-wide imaging of GABAergic neurons in a mouse model of autism (15')
    Fabian Kaiser  (Rotterdam) The role of midbrain inhibition in autism-like behaviours.  (15')

    Session 31: Gut feelings: the role of neuroendocrinological factors in regulating food intake and stress processes
    Chairs: Karlijn Kooij & Frank Meye (Utrecht)

    Alfonso Abizaid  (Ottawa, Canada) Stomaching Stress: how ghrelin is a hormone that is important for coping with chronic stressors (30')
    Karlijn Kooij  (Utrecht) 
    Ghrelin alters dopaminergic and GABAergic ventral tegmental area neuronal signaling during a food-predictive cue (15')

    Jari Berkhout  (Leiden) An integrated single-cell RNA-seq atlas of the mouse hypothalamic paraventricular nucleus links transcriptomic and functional types (15')
    Iris Stoltenborg  (Gothenburg, Sweden) Activation of a ghrelin-responsive neuronal “hunger” ensemble in the dorsomedial hypothalamus is sufficient to impact on food intake, hunger valence evaluation and food motivation in mice (15')
    Susanne la Fleur  (Amsterdam) Chemogenetic activation of D2/A2a-expressing medium spiny neurons in the nucleus accumbens core mirrors the delaying effect of novelty exposure on feeding onset in rats (15')

    Session 32: Epigenetic regulation of fear memory in health and disease
    Chairs: Kübra Gülmez Karaca & Marloes Henckens (Nijmegen)

    Isabelle Mansuy  (Zürich, Switzerland) Chromatin structure as a molecular substrate for traces of past life experiences (30')
    Janina Kupke  (Amsterdam) Hippocampal DNA methylation promotes memory persistence by facilitating systems consolidation and cortical engram stabilisation. (15')
    Ana Carolina Temporao  (Nijmegen) The epicenter of trauma resilience (15')
    Sabine Spijker  (Amsterdam) Memory recall and hippocampal activation in response to subchronic social defeat stress (15')
    Bart Rutten  (Maastricht) Gene-environment interaction and the biological underpinnings of differential susceptibility to the effects of psychological trauma on mental health  (15')

    Session 33: Microglia balancing the neuronal circuits: from physiological regulation to pathogenesis
    Chairs: Mohit Dubey (Amsterdam) & Christian Madry (Berlin, Germany)

    Christian Madry  (Berlin, Germany) Role of microglia in the maturation and modulation of hippocampal networks (30')
    Annika Mordelt  (Nijmegen) Microglia drive neuronal synchrony: a human iPSC-derived triculture approach (15')
    Kieran Higgins  (Amsterdam) A key role for endocannabinoids in microglia-myelin interactions (15')
    Mohit Dubey  (Amsterdam) Microglia-interneuron crosstalk in an Alzheimer mouse model.  (15')
    Giampaolo Milior  (Paris, France) Microglial cells in the human brain: specific motilities, phenotypes and neuronal interactions in development and epilepsy. (15')

    Session 34: Computational models of brain dynamics
    Chairs: Jorge Mejias (Amsterdam) & Arezoo Alizadeh (Nijmegen)

    Fleur Zeldenrust  (Nijmegen) Heterogeneity and non-linearity: how neurons and networks shape information transfer? (15')
    Jorge Mejias  (Amsterdam) Decision making via distributed evidence accumulation in neocortical networks (15')
    Catherine Sibert  (Groningen) Theories of minds: connecting cognitive architectures to brain data (15')
    Renaud Jolivet  (Maastricht) Learning under energetic constraints (15')
    Matin Jafarian  (Delft) Control theory-based analysis for revealing mechanisms of working memory (15')
    Mario Negrello  (Rotterdam) Motor synergies and cerebellar resonances (15')

    Session 35: Human aspects of neuronal development and network activity during health and disease
    Chairs: Nicky Scheefhals (Nijmegen) & Feline Lindhout (Cambridge, United Kingdom)

    Cécile Charrier  (Paris, France) Molecular mechanisms of synaptic development: insights from human-specific genes (30')
    Feline Lindhout  (Cambridge, United Kingdom) Time is of the essence: mechanistic insights into axonal tract expansion in humans (15')
    Nicky Scheefhals  (Nijmegen) Balancing excitation and inhibition: mechanistic insights from human iPSC-derived neuronal models of ASD (15')
    Lara Janssen  (Amsterdam) The genetic background modulates neuronal activity during iNeuron maturation in STXBP1-Related Disorders disease models (15')
    Katrin Linda  (Leuven, Belgium) Sharing is caring – Astrocytes support neuronal autophagy (15')

    16:30 Posters & drinks
    Session 36: Poster session 2

    18:00 Closure DNM 24 & poster prizes

    Molecular mechanisms in neurodegeneration

    Neurodegenerative diseases are characterized by the degeneration of neuronal cells in the brain, causing a wide variety of conditions. With the aging world population, these incurable diseases will only become more prevalent. Recently, the approval of several groundbreaking drugs has shown that the massive effords in the past decades to understand the molecular mechanisms underlying neurodegeneration,have proven to be fruitful. These approved drugs (e.g. Aducanumab for Alzheimer's disease, Tofersen for SOD1-ALS) will pave the way for new, improved drugs that will hopefully be able to cure patients. This session proposes lectures from renowed experts in the field of neurodegeneration, on molecular mechanisms of a variety of neurodegenerative diseases.

    Unlocking the gut-brain connection in aging: from in vitro studies to clinical implications and the role of dietary interventions

    Tackling age-related cognitive decline requires prioritizing preventive strategies. The microbiota-gut-brain axis (MGBA) is an emerging frontier in understanding the intricate interplay between gut health and cognitive function in the elderly. Aging induces changes in gut microbiota composition, leading to e.g., neuroinflammation, altered blood-brain barrier permeability, and amyloid-beta deposition, that increase the risk of cognitive decline. Dietary interventions and specific compounds such as prebiotics targeting the gut show potential for enhancing brain function. Modulating microbiota and its metabolites through the gut-brain axis has demonstrated cognitive improvements in preclinical and clinical studies. Our session gathers experts in MGBA research, exploring its implications for cognitive health in the elderly. Presentations include a keynote on the importance of the gut-brain axis in neurocognition, in vitro co-culture cell models of the gut, blood-brain barrier and neuronal cells, colonic multi-vitamin delivery for neurocognitive enhancement, the impact of prebiotics on cognitive function, and the interplay of the Mediterranean Diet, microbial tryptophan metabolism, and neuroinflammatory status in older adults.

    Computational genomics approaches to study disease mechanisms

    Genomics technologies are becoming increasingly powerful to map the genome structure of individual cell types in health and disease. Consequently, there is a growing demand for computational tools to analyze these complex multi-omics datasets. This session aims to get together a number of prominent Dutch and international researchers who utilize innovative genomics technologies and computational approaches as a means to explore the mechanisms of brain diseases. Dr. Nott aims to decipher how cell type-specific enhancer function drives aging-related disease. Dr. Haghverdi will discuss computational approaches for temporal inferences from high-throughput single-cell (sc) measurements. Dr. Mahfouz will discuss long-read sc-sequencing data to predict exon inclusion in neurons and glia. Charlotte van Dijk will discuss how new computational tools can help to detect novel Alternative polyAdenylation in the context of neurodegeneration. Dr. Lot de Witte will discuss genetic risk factors for neuropsychiatric disorders and microglia. To summarize, the proposed session will bring together a diverse set of speakers in terms of the field of neurogenomics, computational technologies used, career stage and geographic location.

    Microglial functional and neuroanatomical heterogeneity in diseases

    Microglia play a crucial role in maintaining brain homeostasis and are implicated in various neurological challenges. Understanding their functional and neuroanatomical diversity is key to unraveling complex interactions in brain health and disease. This session explores microglial heterogeneity's diverse aspects in diseases, delving into its implications for neurobiology and therapeutics. Keynote speaker Dr. Tay will discuss her research on the heterogeneity, fate, fundamental biological mechanisms, and functions of microglial cells at single-cell resolution in neurodegenerative diseases. Junior speakers will cover topics like microglia in the subventricular zone's role in neurogenesis and tissue repair, circadian regulation of brain immune responses through timed eating, the impact of genetic obesity on microglia and potential links to neuroinflammation and neurodegeneration, and the use of nanotechnology to study and modulate microglial responses. The session aims to provide a comprehensive overview, fostering discussions on novel research and therapeutic avenues in microglial heterogeneity, uniting experts and emerging researchers to advance our understanding of microglia's multifaceted role in brain health and disease.

    Multi-regional perspective of sensorimotor integration in health and disease

    This parallel session aims to provide a comprehensive examination of the complex networks involved in sensorimotor integration, highlighting the integral roles of the basal ganglia, motor cortex, thalamus, and cerebellum. Sensorimotor integration is a fundamental neural process, crucial for the coordination of sensory perception and motor action, and is pivotal in both simple and complex sensorimotor functions. Our objective is to bring together leading experts and rising stars in the field to share their recent work. We will explore the interactions among these key brain regions during sensorimotor tasks. This session will delve into both normal and abnormal functioning of these areas, offering perspectives on how their interplay guides motor control and how dysfunctions can lead to neurological disorders, or how brain damage causes brain dysfunction. The interdisciplinary lineup of the panel speakers, ranging from cellular neuroscience to clinical linguistics, ensures knowledge exchange to a board audience.

    Human cell modelling of human brain development and associated disorders

    Establishing the molecular and cellular mechanisms that control human brain development and function is essential to understanding mental health. It is now becoming clear that the much higher degree of complexity to the formation and organisation of the human brain is opening significant knowledge gaps. These can be closed by recent advances human induced pluripotent stem cells (iPSC) technology to study and manipulate the human cellular components in vitro. Methods to reprogramme somatic cells into induced pluripotent stem cells (iPSC) and then into neuronal and glial cells are now well-established. However, a healthy brain is more than just this and regulation of how and when the cells interact and connect. We aim to explore new approaches and opportunities to model and manipulate these processes. The session will include use of single cell 'omics techniques that can track developmental trajectories of developing neural cells and reveal differences associated with brain disorders; 3-Dimensional interactions that are required for multicellular brain development; and the functional properties of neuronal networks within these cellular ensembles. In combination these will reveal the molecular details of the human brain function.

    Neurological biomarkers of cancer-related cognitive impairment and fatigue: connecting cancer, (neuro)inflammation and behavior

    Advances in cancer treatment boost survival rates, yet survivors of cancers face rising prevalence of cancer-related cognitive impairment (CRCI) and fatigue (CRF). This societal paradox has led to suggestions that cancer and its treatment exert neurotoxic effects, including cell-senescence, neuroinflammation and disruptions in neural functioning. This symposium brings together researchers working at the intersection of oncology, micro-biology, cognitive neurosciences and epidemiology, to addresses the timely topic to comprehend the intricate neuro-immune pathways through which cancer and its treatment impacts brain function. The integration of these disciplines stands at the forefront of advancing research in a broader range of neurological disorders. By fostering this collaboration, we aim to catalyze the development of innovative prevention and treatment strategies, relevant for the overall well-being of cancer survivors and beyond.

    Modulation of the axon initial segment as a remote control of neuronal activity

    It has only recently become evident that the axon initial segment (AIS), a crucial domain in the proximal axon, plays a powerful role in regulating the output activity of neurons. While our knowledge of proteins present at the AIS is increasing, how the AIS structure and function is regulated by neural activity remains a central question in the field. In this symposium we will explore this question by bringing together experts in cell biology of the AIS and neuronal physiology to understand how a central, yet overlooked aspect of neuronal plasticity, controls neuronal activity in health and disease. Using different yet complementary models our speakers will discuss recent work on the extrinsic and intrinsic mechanisms controlling AIS plasticity. Notably, we will show how developmental processes and activity levels in neuronal networks regulate AIS function in vivo and emphasize how the use of super-resolution microscopy, new live-imaging approaches and proteomics allow to decipher the structure and function of the AIS. Finally, we will present exciting new data on human induced pluripotent stem cell (iPSCs) neurons derived from patients providing insights into the role of AIS maladaptive plasticity in human diseases.

    Neurobiological substrates of sex differences in anxiety and stress

    Stress-related disorders are much more prevalent in women than men. Still, preclinical animal work investigating the mechanistic underpinnings of stress-related psychopathology often focuses on the male brain only, illustrating the need for sex-specific research. Studies comparing females and males often show both behavioural and neurobiological differences, suggesting the existence of sex-specific neuronal and molecular networks. Given the high prevalence of stress-related disorders, a fundemental understanding of the sex-specific mechanistic underpinnings, is essential. In this session, we will discuss research from several laboratories that are working on sex differences in anxiety and stress and the underlying neurobiological substrates in rodents.

    Wrapping the brain: oligodendrogenesis and myelination in health and disease

    The contribution of white matter abnormalities to neuronal dysfunction is hardly studied on molecular and cellular levels. Myelination undergoes many changes throughout development and defects in myelination have consistently been implicated in various neuropsychiatric disorders. Insight these processes is crucial for understanding the mechanisms underlying these pathologies. Advancements in fundamental and imaging technologies, such as oligodendrocyte organoids and diffusion tensor imaging, empower researchers to non-invasively explore myelination dynamics in living cells and brains. This opens new avenues for studying these processes in brain pathologies. Recent studies underscore the plasticity of oligodendrocytes and their potential for regeneration, fueling interest in therapies that stimulate oligodendrogenesis for neurorepair. This focus on oligodendrogenesis aligns with a broader trend in neuroscience, emphasizing a deep understanding of intricate cellular and molecular processes governing brain function. Targeting oligodendrocyte function carries transformative potential for therapeutic strategies across various neurological disorders, amplifying the promise of this research field within the current scientific landscape.

    Bidirectional insights: how neuroscience and machine learning inform and transform each other

    The convergence of machine learning and neuroscience stands at the vanguard of scientific inquiry, largely due to unparalleled technological advancements that enable the mining of complex neural data sets. This symbiotic relationship not only deepens our understanding of neural processes but also drives the evolution of sophisticated artificial intelligence models. In our planned session, we delve into the significant ways in which deep neural networks can elucidate the emergence and physiological functions of the visual system. We will demonstrate how biological insights inspire advanced computational approaches. Additionally, we aim to illustrate how integrating machine learning frameworks can leverage fMRI data to predict cognitive decline. Our session will also illuminate how deep neural networks are instrumental in identifying diverse sleep states across rodents and humans, uncovering translational and clinical applications that bridge basic research and therapeutic strategies. Another area of focus will be the concept of causality within neural networks, drawing parallels to causality in biological systems. By doing so, we aim to foster a richer understanding of neural mechanisms and their implications for machine learning.

    Identifying novel neurobiological targets for the development of innovative treatment strategies for substance use disorders

    Substance use is a leading cause of disability and death worldwide. Despite the existence of a variety of evidence-based (behavioral and pharmacological) treatments for substance use disorders, relapse rates following treatment remain high and individual differences in treatment response are often overlooked. This underlines the importance of developing more effective and personalized treatment options. Having more treatment options available allows for the development of more personalized treatment strategies, tailored to the individuals’ needs and wishes. Ultimately, this will lead to a reduction of recurrent help seeking and consequently less pressure on healthcare systems. In this session we will discuss the potential of various innovative treatment strategies, including psychedelic-assisted therapy, deep transcranial magnetic stimulation, heart rate variability biofeedback and interventions that target the reconsolidation of addiction-related memories. This session will be concluded with a comprehensive overview of potential sex and gender-differences in the working mechanisms of these novel interventions.

    Keynote Lecture

    Interneuron dynamics during brain development and maturation in health and disease

    Interneurons, the inhibitory cells of the brain display the fundamental function of regulating brain development and cognitive states. To be integrated into specific neuronal circuits, interneurons perform dynamic displacements in the brain before reaching the final destination. Interneurons are also one of the most diverse neuronal cell populations and interneuron diversity is related to complexity in function. It is thus an open question in the field how and when does interneuron diversity emerge and whether it is more driven by intrinsic or extrinsic factors. It is essential to combine different tools and scientific approaches to deeply characterize the intrinsic programs involved in interneuron connectivity while unveiling how the interaction with the extracellular space contributes to their final integration into circuits. In this parallel session we will adress the novel conceptual and technical approaches that are being used in the field to answering to these fundamental questions. This knowledge has high translational potential to tackle complex neurodevelopmental disorders that have deep roots in developmetal stages where interneuron diversity is being established.

    Gut feelings: the role of neuroendocrinological factors in regulating food intake and stress processes

    Organisms must swiftly respond to environmental threats for survival and look for food when hungry. This response is partially orchestrated by the neuroendocrine system, crucially informing the brain. Understanding the gut-brain axis, intricately linking food digestion to profound neurological consequences, is progressing rapidly. Stress further influences endocrine factors and subsequent brain responses. Our distinguished speakers will dissect the exciting developments within this important research topic, ranging from cellular to functional brain changes in the context of food intake and stress-related neuroendocrine release. A better understanding of the neuroendocrine system will provide useful insight into therapeutic strategies on binge eating disorder, obesity and anxiety disorders.

    Epigenetic regulation of fear memory in health and disease

    Epigenetic mechanisms, such as DNA methylation, histone modifications or higher-order chromatin organization, have emerged as critical regulators of memory processing. Dynamic modulation of these epigenetic mechanisms upon neuronal activity ensures an optimal genomic response required for the consolidation of long-term memory. Interestingly, recent evidence suggests that alterations in epigenetic mechanisms may also be involved in the aberrant processing of trauma memory and may pose a risk for developing PTSD(-like) symptomatology. In this symposium, we will discuss recent findings on the contribution of epigenetic mechanisms regulating the storage of fear memories and how this can go wrong in disease. We cover animal working allowing for unprecedented precision in their studies of how epigenetic mechanisms modulate memory processing in the actual cells that store the memory (i.e., the memory engram). To link this to human-relevant pathology, we additionally present work investigating deviations in epigenetic regulators in clinical populations (PTSD patients). In doing so, we aim to further the understanding of epigenetic mechanisms within healthy and pathological forms of fear memory storage in mice to humans.

    Microglia balancing the neuronal circuits: from physiological regulation to pathogenesis

    In recent years, microglia research has become a pivotal and rapidly evolving field in neuroscience. This surge in interest is largely due to an expanding understanding of the multifaceted roles these cells play in brain function and development. Traditionally recognized as the brain's innate macrophages, microglia are now acknowledged for their significant contributions to synaptic interaction, myelination, and network oscillations, profoundly influencing brain development and functionality. This enhanced perspective has opened new avenues in comprehending and potentially therapeutically addressing a variety of neurological disorders, such as Alzheimer's, autism, and schizophrenia, where microglia's involvement is becoming increasingly apparent. Despite recent advancements, significant knowledge gaps remain, especially regarding microglia's pathophysiological interactions with different cell types, such as GABAergic interneurons and myelinating oligodendrocytes in shaping neuronal circuits. Additionally, microglial activity on brain plasticity and network oscillations remain under-explored. In the session, we aim to bring junior and experienced researchers to discuss microglia physiology in both healthy and diseased brains.

    Computational models of brain dynamics

    Computational neuroscience has emerged as a powerful tool for understanding the complex dynamics of neural systems and their implications for cognition. By integrating principles from neuroscience, physics, and computer science, it seeks to simulate and understand the dynamic interactions within neural networks and provide valuable insights into the mechanisms underlying cognitive processes and neurological phenomena. This session will bring together researchers from diverse backgrounds to discuss recent advances in computational approaches to studying neural dynamics. The goal of this session is to provide a forum for discussing recent advances in the field and to promote interdisciplinary collaboration. Topics of interest include, but are not limited to: modeling of neural circuits, analysis of large-scale neural data, machine learning approaches to neural data analysis, and theoretical frameworks for understanding neural dynamics. The session will consist of six invited talks, each of 15 minutes long. The speakers are experts in their respective fields and will provide a comprehensive overview of the latest research in computational neuroscience. Presentations will have room for discussions and questions.

    Human aspects of neuronal development and network activity during health and disease

    The human brain has unique computational capabilities that enable higher cognitive functions, but also increase our susceptibility to neurological disease. These capabilities rely on the capacity of neurons to develop complex morphologies and interconnect through synapses forming intricate networks that dynamically remodel in response to stimuli. Exploring human aspects of neuronal development and functioning has long remained challenging due to limitations of model organisms and earlier in vitro models. Recent advancements in human in vitro brain models remain revolutionary, notably marked by improved methods that reproducibly generate mature neurons and functional networks derived from human stem cells in both 2D cultures and brain organoids. Hidden inside these complex in vitro neuronal networks lie clues to the unique features of human cognition. Moreover, patient stem cell-derived neurons allow for personalized disease modeling, revealing individual genetic and molecular factors underlying neurological disease. In this session, our speakers will share their pioneering results on human aspects of neuronal development and networks, in link with human brain evolution and neurological disorders with potential therapeutic avenues.

    Poster session 2