← Back

Excitability

Topic spotlight
TopicWorld Wide

excitability

Discover seminars, jobs, and research tagged with excitability across World Wide.
78 curated items40 ePosters38 Seminars
Updated 3 months ago
78 items · excitability
78 results
SeminarNeuroscience

Low intensity rTMS: age dependent effects, and mechanisms underlying neural plasticity

Ann Lohof
Sorbonne Université, Institut de Biologie Paris Seine
Sep 18, 2025

Neuroplasticity is essential for the establishment and strengthening of neural circuits. Repetitive transcranial magnetic stimulation (rTMS) is commonly used to modulate cortical excitability and shows promise in the treatment of some neurological disorders. Low intensity magnetic stimulation (LI-rTMS), which does not directly elicit action potentials in the stimulated neurons, have also shown some therapeutic effects, and it is important to determine the biological mechanisms underlying the effects of these low intensity magnetic fields, such as would occur in the regions surrounding the central high-intensity focus of rTMS. Our team has used a focal low-intensity (10mT) magnetic stimulation approach to address some of these questions and to identify cellular mechanisms. I will present several studies from our laboratory, addressing (1) effects of LIrTMS on neuronal activity and excitability ; and (2) neuronal morphology and post-lesion repair. The ensemble of our results indicate that the effects of LI-rTMS depend upon the stimulation pattern, the age of the animal, and the presence of cellular magnetoreceptors.

SeminarNeuroscience

How are the epileptogenesis clocks ticking?

Cristina Reschke
RCSI
Apr 9, 2024

The epileptogenesis process is associated with large-scale changes in gene expression, which contribute to the remodelling of brain networks permanently altering excitability. About 80% of the protein coding genes are under the influence of the circadian rhythms. These are 24-hour endogenous rhythms that determine a large number of daily changes in physiology and behavior in our bodies. In the brain, the master clock regulates a large number of pathways that are important during epileptogenesis and established-epilepsy, such as neurotransmission, synaptic homeostasis, inflammation, blood-brain barrier among others. In-depth mapping of the molecular basis of circadian timing in the brain is key for a complete understanding of the cellular and molecular events connecting genes to phenotypes.

SeminarNeuroscience

Thalamocortical feedback circuits selectively control pyramidal neuron excitability

Anthony Holtmaat
University of Geneva, Switzerland
Apr 9, 2024
SeminarNeuroscienceRecording

Seizure control by electrical stimulation: parameters and mechanisms

Dominique Durand
Case Western
Jan 30, 2024

Seizure suppression by deep brain stimulation (DBS) applies high frequency stimulation (HFS) to grey matter to block seizures. In this presentation, I will present the results of a different method that employs low frequency stimulation (LFS) (1 to 10Hz) of white matter tracts to prevent seizures. The approach has been shown to be effective in the hippocampus by stimulating the ventral and dorsal hippocampal commissure in both animal and human studies respectively for mesial temporal lobe seizures. A similar stimulation paradigm has been shown to be effective at controlling focal cortical seizures in rats with corpus callosum stimulation. This stimulation targets the axons of the corpus callosum innervating the focal zone at low frequencies (5 to 10Hz) and has been shown to significantly reduce both seizure and spike frequency. The mechanisms of this suppression paradigm have been elucidated with in-vitro studies and involve the activation of two long-lasting inhibitory potentials GABAB and sAHP. LFS mechanisms are similar in both hippocampus and cortical brain slices. Additionally, the results show that LFS does not block seizures but rather decreases the excitability of the tissue to prevent seizures. Three methods of seizure suppression, LFS applied to fiber tracts, HFS applied to focal zone and stimulation of the anterior nucleus of the thalamus (ANT) were compared directly in the same animal in an in-vivo epilepsy model. The results indicate that LFS generated a significantly higher level of suppression, indicating LFS of white matter tract could be a useful addition as a stimulation paradigm for the treatment of epilepsy.

SeminarNeuroscience

Sleep deprivation and the human brain: from brain physiology to cognition”

Ali Salehinejad
Leibniz Research Centre for Working Environment & Human Factors, Dortmund, Germany
Aug 28, 2023

Sleep strongly affects synaptic strength, making it critical for cognition, especially learning and memory formation. Whether and how sleep deprivation modulates human brain physiology and cognition is poorly understood. Here we examined how overnight sleep deprivation vs overnight sufficient sleep affects (a) cortical excitability, measured by transcranial magnetic stimulation, (b) inducibility of long-term potentiation (LTP)- and long-term depression (LTD)-like plasticity via transcranial direct current stimulation (tDCS), and (c) learning, memory, and attention. We found that sleep deprivation increases cortical excitability due to enhanced glutamate-related cortical facilitation and decreases and/or reverses GABAergic cortical inhibition. Furthermore, tDCS-induced LTP-like plasticity (anodal) abolishes while the inhibitory LTD-like plasticity (cathodal) converts to excitatory LTP-like plasticity under sleep deprivation. This is associated with increased EEG theta oscillations due to sleep pressure. Motor learning, behavioral counterparts of plasticity, and working memory and attention, which rely on cortical excitability, are also impaired during sleep deprivation. Our study indicates that upscaled brain excitability and altered plasticity, due to sleep deprivation, are associated with impaired cognitive performance. Besides showing how brain physiology and cognition undergo changes (from neurophysiology to higher-order cognition) under sleep pressure, the findings have implications for variability and optimal application of noninvasive brain stimulation.

SeminarNeuroscienceRecording

Glycolysis regulates neuronal excitability via lactate receptor, HCA1R

Daria Skwarzynska
University of Virginia
May 17, 2023
SeminarNeuroscienceRecording

Feedback control in the nervous system: from cells and circuits to behaviour

Timothy O'Leary
Department of Engineering, University of Cambridge
May 15, 2023

The nervous system is fundamentally a closed loop control device: the output of actions continually influences the internal state and subsequent actions. This is true at the single cell and even the molecular level, where “actions” take the form of signals that are fed back to achieve a variety of functions, including homeostasis, excitability and various kinds of multistability that allow switching and storage of memory. It is also true at the behavioural level, where an animal’s motor actions directly influence sensory input on short timescales, and higher level information about goals and intended actions are continually updated on the basis of current and past actions. Studying the brain in a closed loop setting requires a multidisciplinary approach, leveraging engineering and theory as well as advances in measuring and manipulating the nervous system. I will describe our recent attempts to achieve this fusion of approaches at multiple levels in the nervous system, from synaptic signalling to closed loop brain machine interfaces.

SeminarNeuroscienceRecording

Redox and mitochondrial dysregulation in epilepsy

Manisha Patel
University of Colorado
Sep 20, 2022

Epileptic seizures render the brain uniquely dependent on energy producing pathways. Studies in our laboratory have been focused on the role of redox processes and mitochondria in the context of abnormal neuronal excitability associated with epilepsy. We have shown that that status epilepticus (SE) alters mitochondrial and cellular redox status, energetics and function and conversely, that reactive oxygen species and resultant dysfunction can lead to chronic epilepsy. Oxidative stress and neuroinflammatory pathways have considerable crosstalk and targeting redox processes has recently been shown to control neuroinflammation and excitability. Understanding the role of metabolic and redox processes can enable the development of novel therapeutics to control epilepsy and/or its comorbidities.

SeminarNeuroscience

The role of astroglia-neuron interactions in generation and spread of seizures

Emre Yaksi
Kavli Institute for Systems Neuroscience, Norwegian University of Science and technology
Jul 5, 2022

Astroglia-neuron interactions are involved in multiple processes, regulating development, excitability and connectivity of neural circuits. Accumulating number of evidences highlight a direct connection between aberrant astroglial genetics and physiology in various forms of epilepsies. Using zebrafish seizure models, we showed that neurons and astroglia follow different spatiotemporal dynamics during transitions from pre-ictal to ictal activity. We observed that during pre-ictal period neurons exhibit local synchrony and low level of activity, whereas astroglia exhibit global synchrony and high-level of calcium signals that are anti correlated with neural activity. Instead, generalized seizures are marked by a massive release of astroglial glutamate release as well as a drastic increase of astroglia and neuronal activity and synchrony across the entire brain. Knocking out astroglial glutamate transporters leads to recurrent spontaneous generalized seizures accompanied with massive astroglial glutamate release. We are currently using a combination of genetic and pharmacological approaches to perturb astroglial glutamate signalling and astroglial gap junctions to further investigate their role in generation and spreading of epileptic seizures across the brain.

SeminarNeuroscience

Malignant synaptic plasticity in pediatric high-grade gliomas

Kathryn Taylor
Stanford
May 24, 2022

Pediatric high-grade gliomas (pHGG) are a devastating group of diseases that urgently require novel therapeutic options. We have previously demonstrated that pHGGs directly synapse onto neurons and the subsequent tumor cell depolarization, mediated by calcium-permeable AMPA channels, promotes their proliferation. The regulatory mechanisms governing these postsynaptic connections are unknown. Here, we investigated the role of BDNF-TrkB signaling in modulating the plasticity of the malignant synapse. BDNF ligand activation of its canonical receptor, TrkB (which is encoded for by the gene NTRK2), has been shown to be one important modulator of synaptic regulation in the normal setting. Electrophysiological recordings of glioma cell membrane properties, in response to acute neurotransmitter stimulation, demonstrate in an inward current resembling AMPA receptor (AMPAR) mediated excitatory neurotransmission. Extracellular BDNF increases the amplitude of this glutamate-induced tumor cell depolarization and this effect is abrogated in NTRK2 knockout glioma cells. Upon examining tumor cell excitability using in situ calcium imaging, we found that BDNF increases the intensity of glutamate-evoked calcium transients in GCaMP6s expressing glioma cells. Western blot analysis indicates the tumors AMPAR properties are altered downstream of BDNF induced TrkB activation in glioma. Cell membrane protein capture (via biotinylation) and live imaging of pH sensitive GFP-tagged AMPAR subunits demonstrate an increase of calcium permeable channels at the tumors postsynaptic membrane in response to BDNF. We find that BDNF-TrkB signaling promotes neuron-to-glioma synaptogenesis as measured by high-resolution confocal and electron microscopy in culture and tumor xenografts. Our analysis of published pHGG transcriptomic datasets, together with brain slice conditioned medium experiments in culture, indicates the tumor microenvironment as the chief source of BDNF ligand. Disruption of the BDNF-TrkB pathway in patient-derived orthotopic glioma xenograft models, both genetically and pharmacologically, results in an increased overall survival and reduced tumor proliferation rate. These findings suggest that gliomas leverage normal mechanisms of plasticity to modulate the excitatory channels involved in synaptic neurotransmission and they reveal the potential to target the regulatory components of glioma circuit dynamics as a therapeutic strategy for these lethal cancers.

SeminarNeuroscience

MicroRNAs as targets in the epilepsies: hits, misses and complexes

David Henshall
The Royal College of Surgeons in Ireland
May 3, 2022

MicroRNAs are small noncoding RNAs that provide a critical layer of gene expression control. Individual microRNAs variably exert effects across networks of genes via sequence-specific binding to mRNAs, fine-tuning protein levels. This helps coordinate the timing and specification of cell fate transitions during brain development and maintains neural circuit function and plasticity by activity-dependent (re)shaping of synapses and the levels of neurotransmitter components. MicroRNA levels have been found to be altered in tissue from the epileptogenic zone resected from adults with drug-resistant focal epilepsy and this has driven efforts to explore their therapeutic potential, in particular using antisense oligonucleotide (ASOs) inhibitors termed antimirs. Here, we review the molecular mechanisms by which microRNAs control brain excitability and the latest progress towards a microRNA-based treatment for temporal lobe epilepsy. We also look at whether microRNA-based approaches could be used to treat genetic epilepsies, correcting individual genes or dysregulated pathways. Finally, we look at how cells have evolved to maximise the efficiency of the microRNA system via RNA editing, where single base changes is capable of altering the repertoire of genes under the control of a single microRNA. The findings improve our understanding of the molecular landscape of the epileptic brain and may lead to new therapies.

SeminarNeuroscienceRecording

How does the metabolically-expensive mammalian brain adapt to food scarcity?

Zahid Padamsey
Rochefort lab, University of Edinburgh
Feb 22, 2022

Information processing is energetically expensive. In the mammalian brain, it is unclear how information coding and energy usage are regulated during food scarcity. I addressed this in the visual cortex of awake mice using whole-cell recordings and two-photon imaging to monitor layer 2/3 neuronal activity and ATP usage. I found that food restriction reduced synaptic ATP usage by 29% through a decrease in AMPA receptor conductance. Neuronal excitability was nonetheless preserved by a compensatory increase in input resistance and a depolarized resting membrane potential. Consequently, neurons spiked at similar rates as controls, but spent less ATP on underlying excitatory currents. This energy-saving strategy had a cost since it amplified the variability of visually-evoked subthreshold responses, leading to a 32% broadening in orientation tuning and impaired fine visual discrimination. This reduction in coding precision was associated with reduced levels of the fat mass-regulated hormone leptin and was restored by exogenous leptin supplementation. These findings reveal novel mechanisms that dynamically regulate energy usage and coding precision in neocortex.

SeminarNeuroscienceRecording

Metabolic spikes: from rogue electrons to Parkinson's

Chaitanya Chintaluri
Vogels Lab, IST Austria
Feb 22, 2022

Conventionally, neurons are thought to be cellular units that process synaptic inputs into synaptic spikes. However, it is well known that neurons can also spike spontaneously and display a rich repertoire of firing properties with no apparent functional relevance e.g. in in vitro cortical slice preparations. In this talk, I will propose a hypothesis according to which intrinsic excitability in neurons may be a survival mechanism to minimize toxic byproducts of the cell’s energy metabolism. In neurons, this toxicity can arise when mitochondrial ATP production stalls due to limited ADP. Under these conditions, electrons deviate from the electron transport chain to produce reactive oxygen species, disrupting many cellular processes and challenging cell survival. To mitigate this, neurons may engage in ADP-producing metabolic spikes. I will explore the validity of this hypothesis using computational models that illustrate the implications of synaptic and metabolic spiking, especially in the context of substantia nigra pars compacta dopaminergic neurons and their degeneration in Parkinson's disease.

SeminarNeuroscience

Stress deceleration theory: chronic adolescent stress exposure results in decelerated neurobehavioral maturation

Kshitij Jadhav
University of Cambridge
Jan 18, 2022

Normative development in adolescence indicates that the prefrontal cortex is still under development thereby unable to exert efficient top-down inhibitory control on subcortical regions such as the basolateral amygdala and the nucleus accumbens. This imbalance in the developmental trajectory between cortical and subcortical regions is implicated in expression of the prototypical impulsive, compulsive, reward seeking and risk-taking adolescent behavior. Here we demonstrate that a chronic mild unpredictable stress procedure during adolescence in male Wistar rats arrests the normal behavioral maturation such that they continue to express adolescent-like impulsive, hyperactive, and compulsive behaviors into late adulthood. This arrest in behavioral maturation is associated with the hypoexcitability of prelimbic cortex (PLC) pyramidal neurons and reduced PLC-mediated synaptic glutamatergic control of BLA and nucleus accumbens core (NAcC) neurons that lasts late into adulthood. At the same time stress exposure in adolescence results in the hyperexcitability of the BLA pyramidal neurons sending stronger glutamatergic projections to the NAcC. Chemogenetic reversal of the PLC hypoexcitability decreased compulsivity and improved the expression of goal-directed behavior in rats exposed to stress during adolescence, suggesting a causal role for PLC hypoexcitability in this stress-induced arrested behavioral development. (https://www.biorxiv.org/content/10.1101/2021.11.21.469381v1.abstract)

SeminarNeuroscienceRecording

The GluN2A Subunit of the NMDA Receptor and Parvalbumin Interneurons: A Possible Role in Interneuron Development

Steve Traynelis & Chad Camp
Emory University School of Medicine
Jan 18, 2022

N-methyl-D-aspartate receptors (NMDARs) are excitatory glutamate-gated ion channels that are expressed throughout the central nervous system. NMDARs mediate calcium entry into cells, and are involved in a host of neurological functions. The GluN2A subunit, encoded by the GRIN2A gene, is expressed by both excitatory and inhibitory neurons, with well described roles in pyramidal cells. By using Grin2a knockout mice, we show that the loss of GluN2A signaling impacts parvalbumin-positive (PV) GABAergic interneuron function in hippocampus. Grin2a knockout mice have 33% more PV cells in CA1 compared to wild type but similar cholecystokinin-positive cell density. Immunohistochemistry and electrophysiological recordings show that excess PV cells do eventually incorporate into the hippocampal network and participate in phasic inhibition. Although the morphology of Grin2a knockout PV cells is unaffected, excitability and action-potential firing properties show age-dependent alterations. Preadolescent (P20-25) PV cells have an increased input resistance, longer membrane time constant, longer action-potential half-width, a lower current threshold for depolarization-induced block of action-potential firing, and a decrease in peak action-potential firing rate. Each of these measures are corrected in adulthood, reaching wild type levels, suggesting a potential delay of electrophysiological maturation. The circuit and behavioral implications of this age-dependent PV interneuron malfunction are unknown. However, neonatal Grin2a knockout mice are more susceptible to lipopolysaccharide and febrile-induced seizures, consistent with a critical role for early GluN2A signaling in development and maintenance of excitatory-inhibitory balance. These results could provide insights into how loss-of-function GRIN2A human variants generate an epileptic phenotypes.

SeminarNeuroscienceRecording

NMC4 Short Talk: Resilience through diversity: Loss of neuronal heterogeneity in epileptogenic human tissue impairs network resilience to sudden changes in synchrony

Scott Rich
Kremibl Brain Institute
Nov 30, 2021

A myriad of pathological changes associated with epilepsy, including the loss of specific cell types, improper expression of individual ion channels, and synaptic sprouting, can be recast as decreases in cell and circuit heterogeneity. In recent experimental work, we demonstrated that biophysical diversity is a key characteristic of human cortical pyramidal cells, and past theoretical work has shown that neuronal heterogeneity improves a neural circuit’s ability to encode information. Viewed alongside the fact that seizure is an information-poor brain state, these findings motivate the hypothesis that epileptogenesis can be recontextualized as a process where reduction in cellular heterogeneity renders neural circuits less resilient to seizure onset. By comparing whole-cell patch clamp recordings from layer 5 (L5) human cortical pyramidal neurons from epileptogenic and non-epileptogenic tissue, we present the first direct experimental evidence that a significant reduction in neural heterogeneity accompanies epilepsy. We directly implement experimentally-obtained heterogeneity levels in cortical excitatory-inhibitory (E-I) stochastic spiking network models. Low heterogeneity networks display unique dynamics typified by a sudden transition into a hyper-active and synchronous state paralleling ictogenesis. Mean-field analysis reveals a distinct mathematical structure in these networks distinguished by multi-stability. Furthermore, the mathematically characterized linearizing effect of heterogeneity on input-output response functions explains the counter-intuitive experimentally observed reduction in single-cell excitability in epileptogenic neurons. This joint experimental, computational, and mathematical study showcases that decreased neuronal heterogeneity exists in epileptogenic human cortical tissue, that this difference yields dynamical changes in neural networks paralleling ictogenesis, and that there is a fundamental explanation for these dynamics based in mathematically characterized effects of heterogeneity. These interdisciplinary results provide convincing evidence that biophysical diversity imbues neural circuits with resilience to seizure and a new lens through which to view epilepsy, the most common serious neurological disorder in the world, that could reveal new targets for clinical treatment.

SeminarNeuroscience

Selectively Silencing Nociceptor Sensory Neurons

Clifford J. Woolf
Harvard Medical School
Nov 17, 2021

Local anesthetics decrease the excitability of all neurons by blocking voltage-gated sodium channels non-selectively. We have developed a technology to silence only those sensory neurons – the nociceptors – that trigger pain, itch, and cough. I will tell you why and how we devised the strategy, the way we showed that it works, and will also discuss its implications for treating multiple human disorders.

SeminarNeuroscience

The influence of menstrual cycle on the indices of cortical excitability

Vladimir Djurdjevic
HSE University
Nov 17, 2021

Menstruation is a normal physiological process in women occurring as a result of changes in two ovarian produced hormones – estrogen and progesterone. As a result of these fluctuations, women experience different symptoms in their bodies – their immune system changes (Sekigawa et al, 2004), there are changes in their cardiovascular and digestive system (Millikan, 2006), as well as skin (Hall and Phillips, 2005). But these hormone fluctuations produce major changes in their behavioral pattern as well causing: anxiety, sadness, heightened irritability and anger (Severino and Moline, 1995) which is usually classified as premenstrual syndrome (PMS). In some cases these symptoms severely impair women’s lives and professional help is required. The official diagnosis according to DSM-5 (2013) is premenstrual dysphoric disorder (PMDD). Despite its ubiquitous presence the origins of PMS and PMDD are poorly understood. Some efforts to understand the underlying brain state during the menstruation cycle were performed by using TMS (Smith et al, 1999; 2002; 2003; Inghilleri et al, 2004; Hausmann et al, 2006). But all of these experiments suffer from major shortcomings - no control groups and small number of subjects. Our plan is to address all of these shortcomings and make this the biggest (to our knowledge) experiment of its kind which will, hopefully, provide us with some much needed answers.

SeminarNeuroscienceRecording

Converging mechanisms of epileptogenesis after brain injury

Viji Santhakumar
University of California, Riverside
Oct 5, 2021

Traumatic brain injury (TBI), a leading cause of acquired epilepsy, results in primary cellular injury as well as secondary neurophysiological and inflammatory responses which contribute to epileptogenesis. I will present our recent studies identifying a role for neuro-immune interactions, specifically, the innate immune receptor Toll-like receptor 4 (TLR4), in enhancing network excitability and cell loss in hippocampal dentate gyrus early after concussive brain injury. I will describe results indicating that the transient post-traumatic increases in dentate neurogenesis which occurs during the same early post-injury period augments dentate network excitability and epileptogenesis. I will provide evidence for the beneficial effects of targeting TLR4 and neurogenesis early after brain injury in limiting epileptogenesis. We will discuss potential mechanisms for convergence of the post-traumatic neuro-immune and neurogenic changes and the implications for therapies to reduce neurological deficits and epilepsy after brain injury.

SeminarNeuroscienceRecording

Neocortex saves energy by reducing coding precision during food scarcity

Nathalie Rochefort
University of Edinburgh
Sep 26, 2021

Information processing is energetically expensive. In the mammalian brain, it is unclear how information coding and energy usage are regulated during food scarcity. We addressed this in the visual cortex of awake mice using whole-cell patch clamp recordings and two-photon imaging to monitor layer 2/3 neuronal activity and ATP usage. We found that food restriction resulted in energy savings through a decrease in AMPA receptor conductance, reducing synaptic ATP usage by 29%. Neuronal excitability was nonetheless preserved by a compensatory increase in input resistance and a depolarized resting membrane potential. Consequently, neurons spiked at similar rates as controls, but spent less ATP on underlying excitatory currents. This energy-saving strategy had a cost since it amplified the variability of visually-evoked subthreshold responses, leading to a 32% broadening in orientation tuning and impaired fine visual discrimination. These findings reveal novel mechanisms that dynamically regulate energy usage and coding precision in neocortex.

SeminarNeuroscience

Selectively Silencing Nociceptor Sensory Neurons

Clifford J. Woolf
Harvard Medical School
Sep 22, 2021

Local anesthetics decrease the excitability of all neurons by blocking voltage-gated sodium channels non-selectively. We have developed a technology to silence only those sensory neurons – the nociceptors – that trigger pain, itch, and cough. I will tell you why and how we devised the strategy, the way we showed that it works, and will also discuss its implications for treating multiple human disorders.

SeminarNeuroscienceRecording

Tapeworm larvae in the brain: cellular mechanisms of epilepsy in neurocysticercosis

Joseph Raimondo
University of Cape Town
Aug 3, 2021

Cerebral infection by the larvae of the cestode, Taenia solium (neurocysticercosis), is thought to be the leading cause of adult-acquired epilepsy worldwide. Despite this, little is known about the cellular mechanisms that underlie seizure development in this condition. In this talk I will present our recent data exploring multiple interactions between cestode larvae, neuroinflammatory processes and network excitability. We find that viable cestode larvae are able to strongly suppress microglial activation and inflammatory cytokine release with consequences for the modulation host neuroinflammatory responses and seizure development in vivo. At the same time, larvae produce and release glutamate, with acute excitatory effects on neuronal circuits. We hope that an improved understanding of epileptogenic mechanisms in neurocysticercosis will one day improve the management of this condition as well as other inflammatory causes of epilepsy.

SeminarNeuroscienceRecording

Acetylcholine modulation of short-term plasticity is critical to reliable long-term plasticity in hippocampal synapses

Rohan Sharma
Suhita lab, Indian Institute of Science Education and Research Pune
Jul 27, 2021

CA3-CA1 synapses in the hippocampus are the initial locus of episodic memory. The action of acetylcholine alters cellular excitability, modifies neuronal networks, and triggers secondary signaling that directly affects long-term plasticity (LTP) (the cellular underpinning of memory). It is therefore considered a critical regulator of learning and memory in the brain. Its action via M4 metabotropic receptors in the presynaptic terminal of the CA3 neurons and M1 metabotropic receptors in the postsynaptic spines of CA1 neurons produce rich dynamics across multiple timescales. We developed a model to describe the activation of postsynaptic M1 receptors that leads to IP3 production from membrane PIP2 molecules. The binding of IP3 to IP3 receptors in the endoplasmic reticulum (ER) ultimately causes calcium release. This calcium release from the ER activates potassium channels like the calcium-activated SK channels and alters different aspects of synaptic signaling. In an independent signaling cascade, M1 receptors also directly suppress SK channels and the voltage-activated KCNQ2/3 channels, enhancing post-synaptic excitability. In the CA3 presynaptic terminal, we model the reduction of the voltage sensitivity of voltage-gated calcium channels (VGCCs) and the resulting suppression of neurotransmitter release by the action of the M4 receptors. Our results show that the reduced initial release probability because of acetylcholine alters short-term plasticity (STP) dynamics. We characterize the dichotomy of suppressing neurotransmitter release from CA3 neurons and the enhanced excitability of the postsynaptic CA1 spine. Mechanisms underlying STP operate over a few seconds, while those responsible for LTP last for hours, and both forms of plasticity have been linked with very distinct functions in the brain. We show that the concurrent suppression of neurotransmitter release and increased sensitivity conserves neurotransmitter vesicles and enhances the reliability in plasticity. Our work establishes a relationship between STP and LTP coordinated by neuromodulation with acetylcholine.

SeminarNeuroscience

Capacitance clamp - artificial capacitance in biological neurons via dynamic clamp

Paul Pfeiffer
Schreiber lab, Humboldt University Berlin, Germany
Jun 9, 2021

A basic time scale in neural dynamics from single cells to the network level is the membrane time constant - set by a neuron’s input resistance and its capacitance. Interestingly, the membrane capacitance appears to be more dynamic than previously assumed with implications for neural function and pathology. Indeed, altered membrane capacitance has been observed in reaction to physiological changes like neural swelling, but also in ageing and Alzheimer's disease. Importantly, according to theory, even small changes of the capacitance can affect neuronal signal processing, e.g. increase network synchronization or facilitate transmission of high frequencies. In experiment, robust methods to modify the capacitance of a neuron have been missing. Here, we present the capacitance clamp - an electrophysiological method for capacitance control based on an unconventional application of the dynamic clamp. In its original form, dynamic clamp mimics additional synaptic or ionic conductances by injecting their respective currents. Whereas a conductance directly governs a current, the membrane capacitance determines how fast the voltage responds to a current. Accordingly, capacitance clamp mimics an altered capacitance by injecting a dynamic current that slows down or speeds up the voltage response (Fig 1 A). For the required dynamic current, the experimenter only has to specify the original cell and the desired target capacitance. In particular, capacitance clamp requires no detailed model of present conductances and thus can be applied in every excitable cell. To validate the capacitance clamp, we performed numerical simulations of the protocol and applied it to modify the capacitance of cultured neurons. First, we simulated capacitance clamp in conductance based neuron models and analysed impedance and firing frequency to verify the altered capacitance. Second, in dentate gyrus granule cells from rats, we could reliably control the capacitance in a range of 75 to 200% of the original capacitance and observed pronounced changes in the shape of the action potentials: increasing the capacitance reduced after-hyperpolarization amplitudes and slowed down repolarization. To conclude, we present a novel tool for electrophysiology: the capacitance clamp provides reliable control over the capacitance of a neuron and thereby opens a new way to study the temporal dynamics of excitable cells.

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

Brief Sensory Deprivation Triggers Cell Type-Specific Structural and Functional Plasticity in Olfactory Bulb Neurons

Li Huang, Joseph Innes, Emily Winson-Bushby
University of Cambridge, PDN
Apr 27, 2021

Can alterations in experience trigger different plastic modifications in neuronal structure and function, and if so, how do they integrate at the cellular level? To address this question, we interrogated circuitry in the mouse olfactory bulb responsible for the earliest steps in odor processing. We induced experience-dependent plasticity in mice of either sex by blocking one nostril for one day, a minimally invasive manipulation that leaves the sensory organ undamaged and is akin to the natural transient blockage suffered during common mild rhinal infections. We found that such brief sensory deprivation produced structural and functional plasticity in one highly specialized bulbar cell type: axon-bearing dopaminergic neurons in the glomerular layer. After 24 h naris occlusion, the axon initial segment (AIS) in bulbar dopaminergic neurons became significantly shorter, a structural modification that was also associated with a decrease in intrinsic excitability. These effects were specific to the AIS-positive dopaminergic subpopulation because no experience-dependent alterations in intrinsic excitability were observed in AIS-negative dopaminergic cells. Moreover, 24 h naris occlusion produced no structural changes at the AIS of bulbar excitatory neurons, mitral/tufted and external tufted cells, nor did it alter their intrinsic excitability. By targeting excitability in one specialized dopaminergic subpopulation, experience-dependent plasticity in early olfactory networks might act to fine-tune sensory processing in the face of continually fluctuating inputs. (https://www.jneurosci.org/content/41/10/2135)

SeminarNeuroscience

The retrotrapezoid nucleus: an integrative and interoceptive hub in neural control of breathing

Douglas A. Bayliss
University of Virginia
Apr 11, 2021

In this presentation, we will discuss the cellular and molecular properties of the retrotrapezoid nucleus (RTN), an integrative and interoceptive control node for the respiratory motor system. We will present the molecular profiling that has allowed definitive identification of a cluster of tonically active neurons that provide a requisite drive to the respiratory central pattern generator (CPG) and other pre-motor neurons. We will discuss the ionic basis for steady pacemaker-like firing, including by a large subthreshold oscillation; and for neuromodulatory influences on RTN activity, including by arousal state-dependent neurotransmitters and CO2/H+. The CO2/H+-dependent modulation of RTN excitability represents the sensory component of a homeostatic system by which the brain regulates breathing to maintain blood gases and tissue pH; it relies on two intrinsic molecular proton detectors, both a proton-activated G protein-coupled receptor (GPR4) and a proton-inhibited background K+ channel (TASK-2). We will also discuss downstream neurotransmitter signaling to the respiratory CPG, focusing especially on a newly-identified peptidergic modulation of the preBötzinger complex that becomes activated following birth and the initiation of air breathing. Finally, we will suggest how the cellular and molecular properties of RTN neurons identified in rodent models may contribute to understanding human respiratory disorders, such as congenital central hypoventilation syndrome (CCHS) and sudden infant death syndrome (SIDS).

SeminarNeuroscience

Digging Deep: Uncovering Hidden Connections Between Epilepsy and Alzheimer’s Disease

Alice Lam
Harvard University
Mar 22, 2021

An emerging hypothesis in the field of Alzheimer’s disease (AD) is that neuronal hyperexcitability and other forms of aberrant network activity play an important role in shaping the clinical course of AD. In this talk, Dr. Lam will highlight the close and bi-directional relationships between epilepsy and AD, noting recent advances in our understanding of this topic spanning from animal models to humans. She will describe recent intracranial electrode recordings in humans that have revealed silent hippocampal epileptiform activity occurring in early stages of AD. Finally, she will discuss machine learning approaches that her laboratory has been developing to non-invasively diagnose and quantify silent hippocampal epileptiform activity from scalp EEG recordings.

SeminarNeuroscienceRecording

Playing fast and loose with glutamate builds healthy circuits in the developing cortex

Chris Dulla
Tufts University
Feb 16, 2021

The construction of cortical circuits requires the precise formation of connections between excitatory and inhibitory neurons during early development. Multiple factors, including neurotransmitters, neuronal activity, and neuronal-glial interactions, shape how these critical circuits form. Disruptions of these early processes can disrupt circuit formation, leading to epilepsy and other neurodevelopmental disorders. Here, I will describe our work into understanding how prolonged post-natal astrocyte development in the cortex creates a permissive window for glutamate signaling that provides tonic activation of developing interneurons through Grin2D NMDA receptors. Experimental disruption of this pathway results in hyperexcitable cortical circuits and human mutations in the Grin2D gene, as well as other related molecules that regulate early life glutamate signaling, are associated with devastating epileptic encephalopathies. We will explore fundamental mechanisms linking early life glutamate signaling and later circuit hyperexcitability, with an emphasis on potential therapeutic interventions aimed at reducing epilepsy and other neurological dysfunction.

SeminarNeuroscience

K+ Channel Gain of Function in Epilepsy, from Currents to Networks

Matthew Weston
University of Vermont
Oct 20, 2020

Recent human gene discovery efforts show that gain-of-function (GOF) variants in the KCNT1gene, which encodes a Na+-activated K+ channel subunit, cause severe epilepsies and other neurodevelopmental disorders. Although the impact of these variants on the biophysical properties of the channels is well characterized, the mechanisms that link channel dysfunction to cellular and network hyperexcitability and human disease are unknown. Furthermore, precision therapies that correct channel biophysics in non-neuronal cells have had limited success in treating human disease, highlighting the need for a deeper understanding of how these variants affect neurons and networks. To address this gap, we developed a new mouse model with a pathogenic human variant knocked into the mouse Kcnt1gene. I will discuss our findings on the in vivo phenotypes of this mouse, focusing on our characterization of epileptiform neural activity using electrophysiology and widefield Ca++imaging. I will also talk about our investigations at the synaptic, cellular, and circuit levels, including the main finding that cortical inhibitory neurons in this model show a reduction in intrinsic excitability and action potential generation. Finally, I will discuss future directions to better understand the mechanisms underlying the cell-type specific effects, as well as the link between the cellular and network level effects of KCNT1 GOF.

SeminarNeuroscienceRecording

Sparks, flames, and inferno: epileptogenesis in the glioblastoma microenvironment

Jeff Noebels
Baylor College of Medicine
Oct 6, 2020

Glioblastoma cells trigger pharmacoresistant seizures that may promote tumor growth and diminish the quality of remaining life. To define the relationship between growth of glial tumors and their neuronal microenvironment, and to identify genomic biomarkers and mechanisms that may point to better prognosis and treatment of drug resistant epilepsy in brain cancer, we are analyzing a new generation of genetically defined CRISPR/in utero electroporation inborn glioblastoma (GBM) tumor models engineered in mice. The molecular pathophysiology of glioblastoma cells and surrounding neurons and untransformed astrocytes are compared at serial stages of tumor development. Initial studies reveal that epileptiform EEG spiking is a very early and reliable preclinical signature of GBM expansion in these mice, followed by rapidly progressive seizures and death within weeks. FACS-sorted transcriptomic analysis of cortical astrocytes reveals the expansion of a subgroup enriched in pro-synaptogenic genes that may drive hyperexcitability, a novel mechanism of epileptogenesis. Using a prototypical GBM IUE model, we systematically define and correlate the earliest appearance of cortical hyperexcitability with progressive cortical tumor cell invasion, including spontaneous episodes of spreading cortical depolarization, innate inflammation, and xCT upregulation in the peritumoral microenvironment. Blocking this glutamate exporter reduces seizure load. We show that the host genome contributes to seizure risk by generating tumors in a monogenic deletion strain (MapT/tau -/-) that raises cortical seizure threshold. We also show that the tumor variant profile determines epilepsy risk. Our genetic dissection approach sets the stage to broadly explore the developmental biology of personalized tumor/host interactions in mice engineered with novel human tumor mutations in specified glial cell lineages.

SeminarNeuroscience

Autism-Associated Shank3 Is Essential for Homeostatic Compensation in Rodent Visual Cortex

Gina Turrigiano
Brandeis University
Jul 20, 2020

Neocortical networks must generate and maintain stable activity patterns despite perturbations induced by learning and experience- dependent plasticity. There is abundant theoretical and experimental evidence that network stability is achieved through homeostatic plasticity mechanisms that adjust synaptic and neuronal properties to stabilize some measure of average activity, and this process has been extensively studied in primary visual cortex (V1), where chronic visual deprivation induces an initial drop in activity and ensemble average firing rates (FRs), but over time activity is restored to baseline despite continued deprivation. Here I discuss recent work from the lab in which we followed this FR homeostasis in individual V1 neurons in freely behaving animals during a prolonged visual deprivation/eye-reopening paradigm. We find that - when FRs are perturbed by manipulating sensory experience - over time they return precisely to a cell-autonomous set-point. Finally, we find that homeostatic plasticity is perturbed in a mouse model of Autism spectrum disorder, and this results in a breakdown of FRH within V1. These data suggest that loss of homeostatic plasticity is one primary cause of excitation/inhibition imbalances in ASD models. Together these studies illuminate the role of stabilizing plasticity mechanisms in the ability of neocortical circuits to recover robust function following challenges to their excitability.

SeminarNeuroscience

Neuronal morphology imposes a tradeoff between stability, accuracy and efficiency of synaptic scaling

Adriano Bellotti
University of Cambridge
Jul 19, 2020

Synaptic scaling is a homeostatic normalization mechanism that preserves relative synaptic strengths by adjusting them with a common factor. This multiplicative change is believed to be critical, since synaptic strengths are involved in learning and memory retention. Further, this homeostatic process is thought to be crucial for neuronal stability, playing a stabilizing role in otherwise runaway Hebbian plasticity [1-3]. Synaptic scaling requires a mechanism to sense total neuron activity and globally adjust synapses to achieve some activity set-point [4]. This process is relatively slow, which places limits on its ability to stabilize network activity [5]. Here we show that this slow response is inevitable in realistic neuronal morphologies. Furthermore, we reveal that global scaling can in fact be a source of instability unless responsiveness or scaling accuracy are sacrificed." "A neuron with tens of thousands of synapses must regulate its own excitability to compensate for changes in input. The time requirement for global feedback can introduce critical phase lags in a neuron’s response to perturbation. The severity of phase lag increases with neuron size. Further, a more expansive morphology worsens cell responsiveness and scaling accuracy, especially in distal regions of the neuron. Local pools of reserve receptors improve efficiency, potentiation, and scaling, but this comes at a cost. Trafficking large quantities of receptors requires time, exacerbating the phase lag and instability. Local homeostatic feedback mitigates instability, but this too comes at the cost of reducing scaling accuracy." "Realization of the phase lag instability requires a unified model of synaptic scaling, regulation, and transport. We present such a model with global and local feedback in realistic neuron morphologies (Fig. 1). This combined model shows that neurons face a tradeoff between stability, accuracy, and efficiency. Global feedback is required for synaptic scaling but favors either system stability or efficiency. Large receptor pools improve scaling accuracy in large morphologies but worsen both stability and efficiency. Local feedback improves the stability-efficiency tradeoff at the cost of scaling accuracy. This project introduces unexplored constraints on neuron size, morphology, and synaptic scaling that are weakened by an interplay between global and local feedback.

SeminarNeuroscience

Cellular/circuit dysfunction in a model of Dravet syndrome - a severe childhood epilepsy

Ethan M. Goldberg, MD, PhD
The Children's Hospital of Philadelphia
Mar 16, 2020

Dravet syndrome is a severe childhood epilepsy due to heterozygous loss-of-function mutation of the gene SCN1A, which encodes the type 1 neuronal voltage gated sodium (Na+) channel alpha-subunit Nav1.1. Prior studies in mouse models of Dravet syndrome (Scn1a+/- mice) at early developmental time points indicate that, in cerebral cortex, Nav1.1 is predominantly expressed in GABAergic interneurons (INs) and, in particular, in parvalbumin-positive fast-spiking basket cells (PV-INs). This has led to a model of Dravet syndrome pathogenesis whereby Nav1.1 mutation leads to preferential IN dysfunction, decreased synaptic inhibition, hyperexcitability, and epilepsy. We found that, at later developmental time points, the intrinsic excitability of PV-INs has essentially normalized, via compensatory reorganization of axonal Na+ channels. Instead, we found persistent and seemingly paradoxical dysfunction of putative disinhibitory INs expressing vasoactive intestinal peptide (VIP-INs). In vivo two-photon calcium imaging in neocortex during temperature-induced seizures in Scn1a+/- mice showed that mean activity of both putative principal cells and PV-INs was higher in Scn1a+/- relative to wild-type controls during quiet wakefulness at baseline and at elevated core body temperature. However, wild-type PV-INs showed a progressive synchronization in response to temperature elevation that was absent in PV-INs from Scn1a+/- mice immediately prior to seizure onset. We suggest that impaired PV-IN synchronization, perhaps via persistent axonal dysfunction, may contribute to the transition to the ictal state during temperature induced seizures in Dravet syndrome.

ePoster

Adaptive probabilistic regression for real-time motor excitability state prediction from human EEG

Lisa Haxel, Jaivardhan Kapoor, Ulf Ziemann, Jakob Macke

Bernstein Conference 2024

ePoster

Effective excitability: a determinant of the network bursting dynamics revealed by parameter invariance

Oleg Vinogradov, Emmanouil Giannakakis, Betül Uysal, Shlomo Ron, Eyal Weinreb, Holger Lerche, Elisha Moses, Anna Levina

Bernstein Conference 2024

ePoster

Migraine mutation of a Na+ channel induces a switch in excitability type and energetically expensive spikes in an experimentally-constrained model of fast-spiking neurons

Leonardo Preuss, Jan-Hendrik Schleimer, Louisiane Lemaire, Susanne Schreiber

Bernstein Conference 2024

ePoster

Frustrated synchronization and excitability in hierarchical-modular brain networks

COSYNE 2022

ePoster

Frustrated synchronization and excitability in hierarchical-modular brain networks

COSYNE 2022

ePoster

Intrinsic neural excitability induces time-dependent overlap of memory engrams

COSYNE 2022

ePoster

Intrinsic neural excitability induces time-dependent overlap of memory engrams

COSYNE 2022

ePoster

Dendritic excitability primarily controls overdispersion

Zachary Friedenberger & Richard Naud

COSYNE 2023

ePoster

Traveling waves modulate neural excitability along the depth of macaque visual cortex

Lihao Yan, Mitchell Morton, Monika Jadi, Anirvan Nandy

COSYNE 2025

ePoster

Acute aerobic exercise at different intensities modulates motor learning performance and cortical excitability in healthy individuals

Hsiao-I Kuo, Jia-Ling Sun, Ming-Hsien Hsieh, Yi-Ting Lin, Michael Nitsche

FENS Forum 2024

ePoster

Alterations in synaptic and excitability properties of basolateral amygdala neurons in APP/PS1 AD mice

Juliana Gonzalez-Sanmiguel, Loreto San Martin, Scarlet Gallegos, Nicolas Riffo-Lepe, Paulina Saavedra, Luis G. Aguayo

FENS Forum 2024

ePoster

Altered dendritic excitability and cell maturation of CA3 pyramidal neurons during development in the Scn2aA263V genetic epilepsy model

Michela Barboni

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

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

Astrocyte activity triggers adaptive myelin plasticity and increased neuronal excitability in the somatosensory cortex following sensory deprivation

Marina Sánchez-Petidier, Elena Fernandez-López, Elena Alonso-Calviño, Claudia Miguel-Quesada, Alba Fernández-González, José Ángel Rodríguez-Alfaro, Marta Zaforas, M Concepción Serrano, Fernando de Castro, Juan Aguilar, Juliana M Rosa

FENS Forum 2024

ePoster

Beyond maternal boundaries: Exploring offspring excitability in response to maternal chronic stress and mirtazapine

Alžbeta Idunková, Lucia Dubiel-Hoppanová, Katarína Ondáčová, Matúš Tomko, Bohumila Jurkovičová-Tarabová, Stanislava Bukatová, Michal Dubovický, Ľubica Lacinová

FENS Forum 2024

ePoster

CACNA1A haploinsufficiency leads to reduced synaptic function and increased intrinsic excitability

Marina Hommersom, Nina Doorn, Sofía Puvogel, Elly Lewerissa, Annika Mordelt, Ummi Ciptasari, Naoki Kogo, Monica Frega, Dirk Schubert, Bart van de Warrenburg, Nael Nadif Kasri, Hans van Bokhoven

FENS Forum 2024

ePoster

Caspr2 autoantibody pathology is mediated by altered excitability of sensory dorsal root ganglia

Margarita Habib, Anna-Lena Wießler, Patrik Greguletz, Kathrin Doppler, Carmen Villmann

FENS Forum 2024

ePoster

Cell-type specific actions of Nogo-A in controlling spatial memory formation by modulating neuronal excitability

Jan Flechtner, Steffen Fricke, Marta Zagrebelsky, Martin Korte

FENS Forum 2024

ePoster

Chemogenetic activation of the locus coeruleus increases hippocampal noradrenaline levels leading to modulation of hippocampal excitability

Sielke Caestecker, Lars Emil Larsen, Paul Boon, Kristl Vonck, Robrecht Raedt

FENS Forum 2024

ePoster

Chemogenetic elevation of hippocampal excitability unmasks latent ASD risks in non-autistic mice differing in hippocampal AMBRA1 expression and/or sex

Margherita De Introna, Paraskevi Krashia, Annamaria Sabetta, Francesca Stabile, Livia La Barbera, Annalisa Nobili, Marcello D’Amelio, Francesco Cecconi, Martine Ammassari-Teule, Annabella Pignataro

FENS Forum 2024

ePoster

Critical dynamics of cortical excitability and hippocampal seizures

Gregory Lepeu, Ellen van Maren, Antoine Adamantidis, Timothée Proix, Maxime Baud

FENS Forum 2024

ePoster

Deciphering the mechanisms underlying auditory hyperexcitability in a genetic mouse model susceptible to audiogenic seizures

Sabrina Mechaussier, Mathilde Gagliardini, Carolina de Campos Pina, Olivier Postal, Typhaine Dupont, Boris Gourevitch, Nicolas Michalski

FENS Forum 2024

ePoster

Dual effect of anandamide and its endogenous precursor 20:4-NAPE on DRG neuronal excitability and nociception

Anirban Bhattacharyya, Daniel Vasconcelos, Diana Spicarova, Jiri Palecek

FENS Forum 2024

ePoster

The early cognitive decline in Alzheimer's disease models is linked to changes in the intrinsic excitability of the prefrontal cortex

Senka Hadzibegovic, Viviana Villicana Munoz, Francesca Bettoni, Melanie Ginger, Liangying Zhu, Olivier Nicole, Bruno Bontempi, Andreas Frick

FENS Forum 2024

ePoster

Early hippocampal hyperexcitability and mitochondrial changes in a transgenic mouse model of dementia with Lewy bodies

Lauren O'Neill, Chun Chen, Bethany Dennis, Gavin Clowry, Fiona LeBeau

FENS Forum 2024

ePoster

Early maturation and hyperexcitability is a shared phenotype of cortical neurons derived from different ASD-associated mutations

Yara Hussein, Utkarsh Tripathi, Ashwani Choudhary, Ritu Nayak, David Peles, Idan Rosh, Tatiana Rabinski, Gad Vatine, Tali Grain-Shkolnik, Shani Stern

FENS Forum 2024

ePoster

Evidence of prodromal neuronal hyperexcitability and neuroinflammation in a rodent model of human alpha-synucleinopathy

Ibtisam Al Musawi, Gavin Clowry, Fiona Lebeau

FENS Forum 2024

ePoster

So excited to see you! Visual object-in-place learning increases neuronal excitability in lateral entorhinal cortex engram cells

Paul Banks, Gareth Barker, Lisa Kinnavane, Clair Booth, Clea Warburton, Zafar Bashir

FENS Forum 2024

ePoster

Executive dysfunction and abnormal hippocampal-prefrontal intrinsic and synaptic excitability in the 3xTg mouse model for Alzheimer’s disease

Grace Cunliffe, Li Yang Tan, Sarah Luo, Jonathan Turner, Sangyong Jung, John Gigg

FENS Forum 2024

ePoster

Exploring neuroinflammation and neuronal Ca2+ excitability in an Alzheimer's disease mouse model

Martina Bedetta, Nelly Redolfi, Annamaria Lia, Nikita Arnst, Elisa Greotti, Simonetta Falzoni, Francesco Di Virgilio, Paola Pizzo

FENS Forum 2024

ePoster

Functional connectivity as biomarker for network hyperexcitability in Alzheimer’s disease

Anne van Nifterick, Zuzanna Zboś, Cornelis Stam, Alida Gouw

FENS Forum 2024

ePoster

Glycine modulates the excitability of medium spiny neurons in the nucleus accumbens through GPR158 activation

Marcello D'Ascenzo, Giuseppa Aceto, Luca Nardella, Simona Nanni, Valeria Pecci, Alessia Bertozzi, Sofia Nutarelli, Maria Teresa Viscomi, Claudia Colussi, Claudio Grassi

FENS Forum 2024

ePoster

Gradual changes in TMS-induced motor excitability are associated with excitation-inhibition balance dynamics

Lisa Haxel, Paolo Belardinelli, Jakob H. Macke, Ulf Ziemann

FENS Forum 2024

ePoster

Infantile ceroid neuro-lipofuscinosis: Linking autophagy, altered chloride homeostasis, and enhanced brain excitability

Melissa Santi, Simone Giubbolini, Giacomo Pasquini, Tommaso Garavaldi, Vinoshene Pillai, Gabriele Nardi, Gian Michele Ratto, Silvia Landi

FENS Forum 2024

ePoster

Increase in corticospinal cell firing despite reduced excitability of individual corticospinal cells: Paradoxical effects in computer model of motor cortex

William Lytton, Donald Doherty, Adam Newton, Thomas Wichmann, Yoland Smith, Liqiang Chen, Hong-Yuan Chu

FENS Forum 2024

ePoster

Investigation and modulation of cortical excitability in awake rhesus macaques with non-invasive transcranial magnetic stimulation and electroencephalography

Anna Padanyi, Balázs Knakker, Evelin Kiefer, Szuhád Khalil, Antonietta Vitális-Kovács, Rafaella Riszt, Judit Zubánné Inkeller, István Hernádi

FENS Forum 2024

ePoster

Intrinsic excitability of anterior to posterior insula (aIC-pIC) projection neurons are differently modified following retrieval of aversive conditioning

Sailendrakumar Kolatt Chandran, Haneen Kayyal, Federica Cruciani, Adonis Yiannakas, Efrat Edry, Kobi Rosenblum

FENS Forum 2024

ePoster

Microcircuit failure in STXBP1 encephalopathy leads to hyperexcitability

Altair Brito dos Santos, Silas D Larsen, Matthijs Verhage, Jakob B Sørensen, Jean-François Perrier

FENS Forum 2024

ePoster

Neonatal CA3 hyperexcitability drives hippocampal epileptogenesis in SCN2A epileptic encephalopathy

Daniil Kirianov, Yana Reva, Katharina Ulrich, Birgit Engeland, Stephan Marguet, Dirk Isbrandt

FENS Forum 2024