Brain Function
brain function
Professor Geoffrey J Goodhill
The Department of Neuroscience at Washington University School of Medicine is seeking a tenure-track investigator at the level of Assistant Professor to develop an innovative research program in Theoretical/Computational Neuroscience. The successful candidate will join a thriving theoretical/computational neuroscience community at Washington University, including the new Center for Theoretical and Computational Neuroscience. In addition, the Department also has world-class research strengths in systems, circuits and behavior, cellular and molecular neuroscience using a variety of animal models including worms, flies, zebrafish, rodents and non-human primates. The Department’s focus on fundamental neuroscience, outstanding research support facilities, and the depth, breadth and collegiality of our culture provide an exceptional environment to launch your independent research program.
Organization of thalamic networks and mechanisms of dysfunction in schizophrenia and autism
Thalamic networks, at the core of thalamocortical and thalamosubcortical communications, underlie processes of perception, attention, memory, emotions, and the sleep-wake cycle, and are disrupted in mental disorders, including schizophrenia and autism. However, the underlying mechanisms of pathology are unknown. I will present novel evidence on key organizational principles, structural, and molecular features of thalamocortical networks, as well as critical thalamic pathway interactions that are likely affected in disorders. This data can facilitate modeling typical and abnormal brain function and can provide the foundation to understand heterogeneous disruption of these networks in sleep disorders, attention deficits, and cognitive and affective impairments in schizophrenia and autism, with important implications for the design of targeted therapeutic interventions
Neural Signal Propagation Atlas of C. elegans
In the age of connectomics, it is increasingly important to understand how the nodes and edges of a brain's anatomical network, or "connectome," gives rise to neural signaling and neural function. I will present the first comprehensive brain-wide cell-resolved causal measurements of how neurons signal to one another in response to stimulation in the nematode C. elegans. I will compare this signal propagation atlas to the worm's known connectome to address fundamental questions of structure and function in the brain.
Alternative Splicing and Isoforms: role in brain function and pathology
Currents of Hope: how noninvasive brain stimulation is reshaping modern psychiatric care; Adapting to diversity: Integrating variability in brain structure and function into personalized / closed-loop non-invasive brain stimulation for substance use disorders
In March we will focus on TMS and host Ghazaleh Soleimani and Colleen Hanlon. The talks will talk place on Thursday, March 28th at noon ET – please be aware that this means 5PM CET since Boston already switched to summer time! Ghazaleh Soleimani, PhD, is a postdoctoral fellow in Dr Hamed Ekhtiari’s lab at the University of Minnesota. She is also the executive director of the International Network of tES/TMS for Addiction Medicine (INTAM). She will discuss “Adapting to diversity: Integrating variability in brain structure and function into personalized / closed-loop non-invasive brain stimulation for substance use disorders”. Colleen Hanlon, PhD, currently serves as a Vice President of Medical Affairs for BrainsWay, a company specializing in medical devices for mental health, including TMS. Colleen previously worked at the Medical University of South Carolina and Wake Forest School of Medicine. She received the International Brain Stimulation Early Career Award in 2023. She will discuss “Currents of Hope: how noninvasive brain stimulation is reshaping modern psychiatric care”. As always, we will also get a glimpse at the “Person behind the science”. Please register va talks.stimulatingbrains.org to receive the (free) Zoom link, subscribe to our newsletter, or follow us on Twitter/X for further updates!
Novel approaches to non-invasive neuromodulation for neuropsychiatric disorders; Effects of deep brain stimulation on brain function in obsessive-compulsive disorder
On Thursday, February 29th, we will host Damiaan Denys and Andrada Neacsiu. The talks will be followed by a shared discussion. You can register via talks.stimulatingbrains.org to receive the (free) Zoom link!
Neuronal population interactions between brain areas
Most brain functions involve interactions among multiple, distinct areas or nuclei. Yet our understanding of how populations of neurons in interconnected brain areas communicate is in its infancy. Using a population approach, we found that interactions between early visual cortical areas (V1 and V2) occur through a low-dimensional bottleneck, termed a communication subspace. In this talk, I will focus on the statistical methods we have developed for studying interactions between brain areas. First, I will describe Delayed Latents Across Groups (DLAG), designed to disentangle concurrent, bi-directional (i.e., feedforward and feedback) interactions between areas. Second, I will describe an extension of DLAG applicable to three or more areas, and demonstrate its utility for studying simultaneous Neuropixels recordings in areas V1, V2, and V3. Our results provide a framework for understanding how neuronal population activity is gated and selectively routed across brain areas.
Trends in NeuroAI - SwiFT: Swin 4D fMRI Transformer
Trends in NeuroAI is a reading group hosted by the MedARC Neuroimaging & AI lab (https://medarc.ai/fmri). Title: SwiFT: Swin 4D fMRI Transformer Abstract: Modeling spatiotemporal brain dynamics from high-dimensional data, such as functional Magnetic Resonance Imaging (fMRI), is a formidable task in neuroscience. Existing approaches for fMRI analysis utilize hand-crafted features, but the process of feature extraction risks losing essential information in fMRI scans. To address this challenge, we present SwiFT (Swin 4D fMRI Transformer), a Swin Transformer architecture that can learn brain dynamics directly from fMRI volumes in a memory and computation-efficient manner. SwiFT achieves this by implementing a 4D window multi-head self-attention mechanism and absolute positional embeddings. We evaluate SwiFT using multiple large-scale resting-state fMRI datasets, including the Human Connectome Project (HCP), Adolescent Brain Cognitive Development (ABCD), and UK Biobank (UKB) datasets, to predict sex, age, and cognitive intelligence. Our experimental outcomes reveal that SwiFT consistently outperforms recent state-of-the-art models. Furthermore, by leveraging its end-to-end learning capability, we show that contrastive loss-based self-supervised pre-training of SwiFT can enhance performance on downstream tasks. Additionally, we employ an explainable AI method to identify the brain regions associated with sex classification. To our knowledge, SwiFT is the first Swin Transformer architecture to process dimensional spatiotemporal brain functional data in an end-to-end fashion. Our work holds substantial potential in facilitating scalable learning of functional brain imaging in neuroscience research by reducing the hurdles associated with applying Transformer models to high-dimensional fMRI. Speaker: Junbeom Kwon is a research associate working in Prof. Jiook Cha’s lab at Seoul National University. Paper link: https://arxiv.org/abs/2307.05916
Virtual Brain Twins for Brain Medicine and Epilepsy
Over the past decade we have demonstrated that the fusion of subject-specific structural information of the human brain with mathematical dynamic models allows building biologically realistic brain network models, which have a predictive value, beyond the explanatory power of each approach independently. The network nodes hold neural population models, which are derived using mean field techniques from statistical physics expressing ensemble activity via collective variables. Our hybrid approach fuses data-driven with forward-modeling-based techniques and has been successfully applied to explain healthy brain function and clinical translation including aging, stroke and epilepsy. Here we illustrate the workflow along the example of epilepsy: we reconstruct personalized connectivity matrices of human epileptic patients using Diffusion Tensor weighted Imaging (DTI). Subsets of brain regions generating seizures in patients with refractory partial epilepsy are referred to as the epileptogenic zone (EZ). During a seizure, paroxysmal activity is not restricted to the EZ, but may recruit other healthy brain regions and propagate activity through large brain networks. The identification of the EZ is crucial for the success of neurosurgery and presents one of the historically difficult questions in clinical neuroscience. The application of latest techniques in Bayesian inference and model inversion, in particular Hamiltonian Monte Carlo, allows the estimation of the EZ, including estimates of confidence and diagnostics of performance of the inference. The example of epilepsy nicely underwrites the predictive value of personalized large-scale brain network models. The workflow of end-to-end modeling is an integral part of the European neuroinformatics platform EBRAINS and enables neuroscientists worldwide to build and estimate personalized virtual brains.
Deleterious, beneficial or both –retrotransposon expression in brain function and dysfunction
Brain network communication: concepts, models and applications
Understanding communication and information processing in nervous systems is a central goal of neuroscience. Over the past two decades, advances in connectomics and network neuroscience have opened new avenues for investigating polysynaptic communication in complex brain networks. Recent work has brought into question the mainstay assumption that connectome signalling occurs exclusively via shortest paths, resulting in a sprawling constellation of alternative network communication models. This Review surveys the latest developments in models of brain network communication. We begin by drawing a conceptual link between the mathematics of graph theory and biological aspects of neural signalling such as transmission delays and metabolic cost. We organize key network communication models and measures into a taxonomy, aimed at helping researchers navigate the growing number of concepts and methods in the literature. The taxonomy highlights the pros, cons and interpretations of different conceptualizations of connectome signalling. We showcase the utility of network communication models as a flexible, interpretable and tractable framework to study brain function by reviewing prominent applications in basic, cognitive and clinical neurosciences. Finally, we provide recommendations to guide the future development, application and validation of network communication models.
In vivo direct imaging of neuronal activity at high temporospatial resolution
Advanced noninvasive neuroimaging methods provide valuable information on the brain function, but they have obvious pros and cons in terms of temporal and spatial resolution. Functional magnetic resonance imaging (fMRI) using blood-oxygenation-level-dependent (BOLD) effect provides good spatial resolution in the order of millimeters, but has a poor temporal resolution in the order of seconds due to slow hemodynamic responses to neuronal activation, providing indirect information on neuronal activity. In contrast, electroencephalography (EEG) and magnetoencephalography (MEG) provide excellent temporal resolution in the millisecond range, but spatial information is limited to centimeter scales. Therefore, there has been a longstanding demand for noninvasive brain imaging methods capable of detecting neuronal activity at both high temporal and spatial resolution. In this talk, I will introduce a novel approach that enables Direct Imaging of Neuronal Activity (DIANA) using MRI that can dynamically image neuronal spiking activity in milliseconds precision, achieved by data acquisition scheme of rapid 2D line scan synchronized with periodically applied functional stimuli. DIANA was demonstrated through in vivo mouse brain imaging on a 9.4T animal scanner during electrical whisker-pad stimulation. DIANA with milliseconds temporal resolution had high correlations with neuronal spike activities, which could also be applied in capturing the sequential propagation of neuronal activity along the thalamocortical pathway of brain networks. In terms of the contrast mechanism, DIANA was almost unaffected by hemodynamic responses, but was subject to changes in membrane potential-associated tissue relaxation times such as T2 relaxation time. DIANA is expected to break new ground in brain science by providing an in-depth understanding of the hierarchical functional organization of the brain, including the spatiotemporal dynamics of neural networks.
Establishment and aging of the neuronal DNA methylation landscape in the hippocampus
The hippocampus is a brain region with key roles in memory formation, cognitive flexibility and emotional control. Yet hippocampal function is impaired severely during aging and in neurodegenerative diseases, and impairments in hippocampal function underlie age-related cognitive decline. Accumulating evidence suggests that the deterioration of the neuron-specific epigenetic landscape during aging contributes to their progressive, age-related dysfunction. For instance, we have recently shown that aging is associated with pronounced alterations of neuronal DNA methylation patterns in the hippocampus. Because neurons are generated mostly during development with limited replacement in the adult brain, they are particularly long-lived cells and have to maintain their cell-type specific gene expression programs life-long in order to preserve brain function. Understanding the epigenetic mechanisms that underlie the establishment and long-term maintenance of neuron-specific gene expression programs, will help us to comprehend the sources and consequences of their age-related deterioration. In this talk, I will present our recent work that investigated the role of DNA methylation in the establishment of neuronal gene expression programs and neuronal function, using adult neurogenesis in the hippocampus as a model. I will then describe the effects of aging on the DNA methylation landscape in the hippocampus and discuss the malleability of the aging neuronal methylome to lifestyle and environmental stimulation.
Myelin Formation and Oligodendrocyte Biology in Epilepsy
Epilepsy is one of the most common neurological diseases according to the World Health Organization (WHO) affecting around 70 million people worldwide [WHO]. Patients who suffer from epilepsy also suffer from a variety of neuro-psychiatric co-morbidities, which they can experience as crippling as the seizure condition itself. Adequate organization of cerebral white matter is utterly important for cognitive development. The failure of integration of neurologic function with cognition is reflected in neuro-psychiatric disease, such as autism spectrum disorder (ASD). However, in epilepsy we know little about the importance of white matter abnormalities in epilepsy-associated co-morbidities. Epilepsy surgery is an important therapy strategy in patients where conventional anti-epileptic drug treatment fails . On histology of the resected brain samples, malformations of cortical development (MCD) are common among the epilepsy surgery population, especially focal cortical dysplasia (FCD) and tuberous sclerosis complex (TSC). Both pathologies are associated with constitutive activation of the mTOR pathway. Interestingly, some type of FCD is morphological similar to TSC cortical tubers including the abnormalities of the white matter. Hypomyelination with lack of myelin-producing cells, the oligodendrocytes, within the lesional area is a striking phenomenon. Impairment of the complex myelination process can have a major impact on brain function. In the worst case leading to distorted or interrupted neurotransmissions. It is still unclear whether the observed myelin pathology in epilepsy surgical specimens is primarily related to the underlying malformation process or is just a secondary phenomenon of recurrent epileptic seizures creating a toxic micro-environment which hampers myelin formation. Interestingly, mTORC1 has been implicated as key signal for myelination, thus, promoting the maturation of oligodendrocytes . These results, however, remain controversial. Regardless of the underlying pathophysiologic mechanism, alterations of myelin dynamics, depending on their severity, are known to be linked to various kinds of developmental disorders or neuropsychiatric manifestations.
Predictive modeling, cortical hierarchy, and their computational implications
Predictive modeling and dimensionality reduction of functional neuroimaging data have provided rich information about the representations and functional architectures of the human brain. While these approaches have been effective in many cases, we will discuss how neglecting the internal dynamics of the brain (e.g., spontaneous activity, global dynamics, effective connectivity) and its underlying computational principles may hinder our progress in understanding and modeling brain functions. By reexamining evidence from our previous and ongoing work, we will propose new hypotheses and directions for research that consider both internal dynamics and the computational principles that may govern brain processes.
Extracting computational mechanisms from neural data using low-rank RNNs
An influential theory in systems neuroscience suggests that brain function can be understood through low-dimensional dynamics [Vyas et al 2020]. However, a challenge in this framework is that a single computational task may involve a range of dynamic processes. To understand which processes are at play in the brain, it is important to use data on neural activity to constrain models. In this study, we present a method for extracting low-dimensional dynamics from data using low-rank recurrent neural networks (lrRNNs), a highly expressive and understandable type of model [Mastrogiuseppe & Ostojic 2018, Dubreuil, Valente et al. 2022]. We first test our approach using synthetic data created from full-rank RNNs that have been trained on various brain tasks. We find that lrRNNs fitted to neural activity allow us to identify the collective computational processes and make new predictions for inactivations in the original RNNs. We then apply our method to data recorded from the prefrontal cortex of primates during a context-dependent decision-making task. Our approach enables us to assign computational roles to the different latent variables and provides a mechanistic model of the recorded dynamics, which can be used to perform in silico experiments like inactivations and provide testable predictions.
Maths, AI and Neuroscience Meeting Stockholm
To understand brain function and develop artificial general intelligence it has become abundantly clear that there should be a close interaction among Neuroscience, machine learning and mathematics. There is a general hope that understanding the brain function will provide us with more powerful machine learning algorithms. On the other hand advances in machine learning are now providing the much needed tools to not only analyse brain activity data but also to design better experiments to expose brain function. Both neuroscience and machine learning explicitly or implicitly deal with high dimensional data and systems. Mathematics can provide powerful new tools to understand and quantify the dynamics of biological and artificial systems as they generate behavior that may be perceived as intelligent.
Myelin Formation and Oligodendrocyte Biology in Epilepsy
Epilepsy is one of the most common neurological diseases according to the World Health Organization (WHO) affecting around 70 million people worldwide [WHO]. Patients who suffer from epilepsy also suffer from a variety of neuro-psychiatric co-morbidities, which they can experience as crippling as the seizure condition itself. Adequate organization of cerebral white matter is utterly important for cognitive development. The failure of integration of neurologic function with cognition is reflected in neuro-psychiatric disease, such as autism spectrum disorder (ASD). However, in epilepsy we know little about the importance of white matter abnormalities in epilepsy-associated co-morbidities. Epilepsy surgery is an important therapy strategy in patients where conventional anti-epileptic drug treatment fails . On histology of the resected brain samples, malformations of cortical development (MCD) are common among the epilepsy surgery population, especially focal cortical dysplasia (FCD) and tuberous sclerosis complex (TSC). Both pathologies are associated with constitutive activation of the mTOR pathway. Interestingly, some type of FCD is morphological similar to TSC cortical tubers including the abnormalities of the white matter. Hypomyelination with lack of myelin-producing cells, the oligodendrocytes, within the lesional area is a striking phenomenon. Impairment of the complex myelination process can have a major impact on brain function. In the worst case leading to distorted or interrupted neurotransmissions. It is still unclear whether the observed myelin pathology in epilepsy surgical specimens is primarily related to the underlying malformation process or is just a secondary phenomenon of recurrent epileptic seizures creating a toxic micro-environment which hampers myelin formation. Interestingly, mTORC1 has been implicated as key signal for myelination, thus, promoting the maturation of oligodendrocytes . These results, however, remain controversial. Regardless of the underlying pathophysiologic mechanism, alterations of myelin dynamics, depending on their severity, are known to be linked to various kinds of developmental disorders or neuropsychiatric manifestations.
Towards multi-system network models for cognitive neuroscience
Artificial neural networks can be useful for studying brain functions. In cognitive neuroscience, recurrent neural networks are often used to model cognitive functions. I will first offer my opinion on what is missing in the classical use of recurrent neural networks. Then I will discuss two lines of ongoing efforts in our group to move beyond the classical recurrent neural networks by studying multi-system neural networks (the talk will focus on two-system networks). These are networks that combine modules for several neural systems, such as vision, audition, prefrontal, hippocampal systems. I will showcase how multi-system networks can potentially be constrained by experimental data in fundamental ways and at scale.
An open-source miniature two-photon microscope for large-scale calcium imaging in freely moving mice
Due to the unsuitability of benchtop imaging for tasks that require unrestrained movement, investigators have tried, for almost two decades, to develop miniature 2P microscopes-2P miniscopes–that can be carried on the head of freely moving animals. In this talk, I would first briefly review the development history of this technique, and then report our latest progress on developing the new generation of 2P miniscopes, MINI2P, that overcomes the limits of previous versions by both meeting requirements for fatigue-free exploratory behavior during extended recording periods and satisfying demands for further increasing the cell yield by an order of magnitude, to thousands of neurons. The performance and reliability of MINI2P are validated by recordings of spatially tuned neurons in three brain regions and in three behavioral assays. All information about MINI2P is open access, with instruction videos, code, and manuals on public repositories, and workshops will be organized to help new users getting started. MINI2P permits large-scale and high-resolution calcium imaging in freely-moving mice, and opens the door to investigating brain functions during unconstrained natural behaviors.
CNStalk: Mapping brain function with ultra-high field MRI
Growing a world-class precision medicine industry
Monash Biomedical Imaging is part of the new $71.2 million Australian Precision Medicine Enterprise (APME) facility, which will deliver large-scale development and manufacturing of precision medicines and theranostic radiopharmaceuticals for industry and research. A key feature of the APME project is a high-energy cyclotron with multiple production clean rooms, which will be located on the Monash Biomedical Imaging (MBI) site in Clayton. This strategic co-location will facilitate radiochemistry, PET and SPECT research and clinical use of theranostic (therapeutic and diagnostic) radioisotopes produced on-site. In this webinar, MBI’s Professor Gary Egan and Dr Maggie Aulsebrook will explain how the APME will secure Australia’s supply of critical radiopharmaceuticals, build a globally competitive Australian manufacturing hub, and train scientists and engineers for the Australian workforce. They will cover the APME’s state-of-the-art 30 MeV and 18-24 MeV cyclotrons and radiochemistry facilities, as well as the services that will be accessible to students, scientists, clinical researchers, and pharmaceutical companies in Australia and around the world. The APME is a collaboration between Monash University, Global Medical Solutions Australia, and Telix Pharmaceuticals. Professor Gary Egan is Director of Monash Biomedical Imaging, Director of the ARC Centre of Excellence for Integrative Brain Function and a Distinguished Professor at the Turner Institute for Brain and Mental Health, Monash University. He is also lead investigator of the Victorian Biomedical Imaging Capability, and Deputy Director of the Australian National Imaging Facility. Dr Maggie Aulsebrook obtained her PhD in Chemistry at Monash University and specialises in the development and clinical translation of radiopharmaceuticals. She has led the development of several investigational radiopharmaceuticals for first-in-human application. Maggie leads the Radiochemistry Platform at Monash Biomedical Imaging.
Neural circuits of visuospatial working memory
One elementary brain function that underlies many of our cognitive behaviors is the ability to maintain parametric information briefly in mind, in the time scale of seconds, to span delays between sensory information and actions. This component of working memory is fragile and quickly degrades with delay length. Under the assumption that behavioral delay-dependencies mark core functions of the working memory system, our goal is to find a neural circuit model that represents their neural mechanisms and apply it to research on working memory deficits in neuropsychiatric disorders. We have constrained computational models of spatial working memory with delay-dependent behavioral effects and with neural recordings in the prefrontal cortex during visuospatial working memory. I will show that a simple bump attractor model with weak inhomogeneities and short-term plasticity mechanisms can link neural data with fine-grained behavioral output in a trial-by-trial basis and account for the main delay-dependent limitations of working memory: precision, cardinal repulsion biases and serial dependence. I will finally present data from participants with neuropsychiatric disorders that suggest that serial dependence in working memory is specifically altered, and I will use the model to infer the possible neural mechanisms affected.
Four questions about brain and behaviour
Tinbergen encouraged ethologists to address animal behaviour by answering four questions, covering physiology, adaptation, phylogeny, and development. This broad approach has implications for neuroscience and psychology, yet, questions about phylogeny are rarely considered in these fields. Here I describe how phylogeny can shed light on our understanding of brain structure and function. Further, I show that we now have or are developing the data and analytical methods necessary to study the natural history of the human mind.
Astroglial modulation of the antidepressant action of deep brain and bright light stimulation
Even if major depression is now the most common of psychiatric disorders, successful antidepressant treatments are still difficult to achieve. Therefore, a better understanding of the mechanisms of action of current antidepressant treatments is needed to ultimately identify new targets and enhance beneficial effects. Given the intimate relationships between astrocytes and neurons at synapses and the ability of astrocytes to "sense" neuronal communication and release gliotransmitters, an attractive hypothesis is emerging stating that the effects of antidepressants on brain function could be, at least in part, modulated by direct influences of astrocytes on neuronal networks. We will present two preclinical studies revealing a permissive role of glia in the antidepressant response: i) Control of the antidepressant-like effects of rat prefrontal cortex Deep Brain Stimulation (DBS) by astroglia, ii) Modulation of antidepressant efficacy of Bright Light Stimulation (BLS) by lateral habenula astroglia. Therefore, it is proposed that an unaltered neuronal-glial system constitutes a major prerequisite to optimize antidepressant efficacy of DBS or BLS. Collectively, these results pave also the way to the development of safer and more effective antidepressant strategies.
The functional connectome across temporal scales
The view of human brain function has drastically shifted over the last decade, owing to the observation that the majority of brain activity is intrinsic rather than driven by external stimuli or cognitive demands. Specifically, all brain regions continuously communicate in spatiotemporally organized patterns that constitute the functional connectome, with consequences for cognition and behavior. In this talk, I will argue that another shift is underway, driven by new insights from synergistic interrogation of the functional connectome using different acquisition methods. The human functional connectome is typically investigated with functional magnetic resonance imaging (fMRI) that relies on the indirect hemodynamic signal, thereby emphasizing very slow connectivity across brain regions. Conversely, more recent methodological advances demonstrate that fast connectivity within the whole-brain connectome can be studied with real-time methods such as electroencephalography (EEG). Our findings show that combining fMRI with scalp or intracranial EEG in humans, especially when recorded concurrently, paints a rich picture of neural communication across the connectome. Specifically, the connectome comprises both fast, oscillation-based connectivity observable with EEG, as well as extremely slow processes best captured by fMRI. While the fast and slow processes share an important degree of spatial organization, these processes unfold in a temporally independent manner. Our observations suggest that fMRI and EEG may be envisaged as capturing distinct aspects of functional connectivity, rather than intermodal measurements of the same phenomenon. Infraslow fluctuation-based and rapid oscillation-based connectivity of various frequency bands constitute multiple dynamic trajectories through a shared state space of discrete connectome configurations. The multitude of flexible trajectories may concurrently enable functional connectivity across multiple independent sets of distributed brain regions.
GeNN
Large-scale numerical simulations of brain circuit models are important for identifying hypotheses on brain functions and testing their consistency and plausibility. Similarly, spiking neural networks are also gaining traction in machine learning with the promise that neuromorphic hardware will eventually make them much more energy efficient than classical ANNs. In this session, we will present the GeNN (GPU-enhanced Neuronal Networks) framework, which aims to facilitate the use of graphics accelerators for computational models of large-scale spiking neuronal networks to address the challenge of efficient simulations. GeNN is an open source library that generates code to accelerate the execution of network simulations on NVIDIA GPUs through a flexible and extensible interface, which does not require in-depth technical knowledge from the users. GeNN was originally developed as a pure C++ and CUDA library but, subsequently, we have added a Python interface and OpenCL backend. We will briefly cover the history and basic philosophy of GeNN and show some simple examples of how it is used and how it interacts with other Open Source frameworks such as Brian2GeNN and PyNN.
Network science and network medicine: New strategies for understanding and treating the biological basis of mental ill-health
The last twenty years have witnessed extraordinarily rapid progress in basic neuroscience, including breakthrough technologies such as optogenetics, and the collection of unprecedented amounts of neuroimaging, genetic and other data relevant to neuroscience and mental health. However, the translation of this progress into improved understanding of brain function and dysfunction has been comparatively slow. As a result, the development of therapeutics for mental health has stagnated too. One central challenge has been to extract meaning from these large, complex, multivariate datasets, which requires a shift towards systems-level mathematical and computational approaches. A second challenge has been reconciling different scales of investigation, from genes and molecules to cells, circuits, tissue, whole-brain, and ultimately behaviour. In this talk I will describe several strands of work using mathematical, statistical, and bioinformatic methods to bridge these gaps. Topics will include: using artificial neural networks to link the organization of large-scale brain connectivity to cognitive function; using multivariate statistical methods to link disease-related changes in brain networks to the underlying biological processes; and using network-based approaches to move from genetic insights towards drug discovey. Finally, I will discuss how simple organisms such as C. elegans can serve to inspire, test, and validate new methods and insights in networks neuroscience.
How sleep contributes to visual perceptual learning
Sleep is crucial for the continuity and development of life. Sleep-related problems can alter brain function, and cause potentially severe psychological and behavioral consequences. However, the role of sleep in our mind and behavior is far from clear. In this talk, I will present our research on how sleep may play a role in visual perceptual learning (VPL) by using simultaneous magnetic resonance spectroscopy and polysomnography in human subjects. We measured the concentrations of neurotransmitters in the early visual areas during sleep and obtained the excitation/inhibition (E/I) ratio which represents the amount of plasticity in the visual system. We found that the E/I ratio significantly increased during NREM sleep while it decreased during REM sleep. The E/I ratio during NREM sleep was correlated with offline performance gains by sleep, while the E/I ratio during REM sleep was correlated with the amount of learning stabilization. These suggest that NREM sleep increases plasticity, while REM sleep decreases it to solidify once enhanced learning. NREM and REM sleep may play complementary roles, reflected by significantly different neurochemical processing, in VPL.
Brain Basics: A peak into the Brain!
My talk will be a ’Neuro 101’ - also called ‘Basics of Neuroscience’. I hope to introduce the field of Neuroscience and give a brief glimpse into the function, history and evolution of the brain. I will guide you through questions such as - What is a brain? What are its basic building blocks and functions?
If we can make computers play chess, why can't we make them see?
If we can make computers play chess and even Jeopardy and Go, then why can't we make them see like us? How does our brain solve the problem of seeing? I will describe some of our recent insights into understanding object recognition in the brain using behavioral, neuronal and computational methods.
Maths, AI and Neuroscience meeting
To understand brain function and develop artificial general intelligence it has become abundantly clear that there should be a close interaction among Neuroscience, machine learning and mathematics. There is a general hope that understanding the brain function will provide us with more powerful machine learning algorithms. On the other hand advances in machine learning are now providing the much needed tools to not only analyse brain activity data but also to design better experiments to expose brain function. Both neuroscience and machine learning explicitly or implicitly deal with high dimensional data and systems. Mathematics can provide powerful new tools to understand and quantify the dynamics of biological and artificial systems as they generate behavior that may be perceived as intelligent. In this meeting we bring together experts from Mathematics, Artificial Intelligence and Neuroscience for a three day long hybrid meeting. We will have talks on mathematical tools in particular Topology to understand high dimensional data, explainable AI, how AI can help neuroscience and to what extent the brain may be using algorithms similar to the ones used in modern machine learning. Finally we will wrap up with a discussion on some aspects of neural hardware that may not have been considered in machine learning.
Monash Biomedical Imaging highlights from 2021 and looking ahead to 2022
Despite the challenges COVID-19 has continued to present, Monash Biomedical Imaging (MBI) has had another outstanding year in terms of publications and scientific output. In this webinar, Professor Gary Egan, Director of MBI, will present an overview of MBI’s achievements during 2021 and outline the biomedical imaging research programs and partnerships in 2022. His presentation will cover: • MBI operational and research achievements during 2021 • Biomedical imaging technology developments and research outcomes during 2021 • Linked laboratories and research teams at MBI • Progress on the development of a cyclotron and precision radiopharmaceutical facility at Clayton • Emerging research opportunities at the Monash Heart Hospital in cardiology and cardiovascular disease. Professor Gary Egan is Director of Monash Biomedical Imaging, Director of the ARC Centre of Excellence for Integrative Brain Function and a Distinguished Professor at the Turner Institute for Brain and Mental Health, Monash University. He is also lead investigator of the Victorian Biomedical Imaging Capability, and Deputy Director of the Australian National Imaging Facility. His substantive body of published work has made a significant impact on the neuroimaging and neuroscience fields. He has sustained success in obtaining significant grants to support his own research and the development of facilities to advance biomedical imaging.
NMC4 Short Talk: Predictive coding is a consequence of energy efficiency in recurrent neural networks
Predictive coding represents a promising framework for understanding brain function, postulating that the brain continuously inhibits predictable sensory input, ensuring a preferential processing of surprising elements. A central aspect of this view on cortical computation is its hierarchical connectivity, involving recurrent message passing between excitatory bottom-up signals and inhibitory top-down feedback. Here we use computational modelling to demonstrate that such architectural hard-wiring is not necessary. Rather, predictive coding is shown to emerge as a consequence of energy efficiency, a fundamental requirement of neural processing. When training recurrent neural networks to minimise their energy consumption while operating in predictive environments, the networks self-organise into prediction and error units with appropriate inhibitory and excitatory interconnections and learn to inhibit predictable sensory input. We demonstrate that prediction units can reliably be identified through biases in their median preactivation, pointing towards a fundamental property of prediction units in the predictive coding framework. Moving beyond the view of purely top-down driven predictions, we demonstrate via virtual lesioning experiments that networks perform predictions on two timescales: fast lateral predictions among sensory units and slower prediction cycles that integrate evidence over time. Our results, which replicate across two separate data sets, suggest that predictive coding can be interpreted as a natural consequence of energy efficiency. More generally, they raise the question which other computational principles of brain function can be understood as a result of physical constraints posed by the brain, opening up a new area of bio-inspired, machine learning-powered neuroscience research.
Advancing Brain-Computer Interfaces by adopting a neural population approach
Brain-computer interfaces (BCIs) have afforded paralysed users “mental control” of computer cursors and robots, and even of electrical stimulators that reanimate their own limbs. Most existing BCIs map the activity of hundreds of motor cortical neurons recorded with implanted electrodes into control signals to drive these devices. Despite these impressive advances, the field is facing a number of challenges that need to be overcome in order for BCIs to become widely used during daily living. In this talk, I will focus on two such challenges: 1) having BCIs that allow performing a broad range of actions; and 2) having BCIs whose performance is robust over long time periods. I will present recent studies from our group in which we apply neuroscientific findings to address both issues. This research is based on an emerging view about how the brain works. Our proposal is that brain function is not based on the independent modulation of the activity of single neurons, but rather on specific population-wide activity patters —which mathematically define a “neural manifold”. I will provide evidence in favour of such a neural manifold view of brain function, and illustrate how advances in systems neuroscience may be critical for the clinical success of BCIs.
Generative models of brain function: Inference, networks, and mechanisms
This talk will focus on the generative modelling of resting state time series or endogenous neuronal activity. I will survey developments in modelling distributed neuronal fluctuations – spectral dynamic causal modelling (DCM) for functional MRI – and how this modelling rests upon functional connectivity. The dynamics of brain connectivity has recently attracted a lot of attention among brain mappers. I will also show a novel method to identify dynamic effective connectivity using spectral DCM. Further, I will summarise the development of the next generation of DCMs towards large-scale, whole-brain schemes which are computationally inexpensive, to the other extreme of the development using more sophisticated and biophysically detailed generative models based on the canonical microcircuits.
Activity dependent myelination: a mechanism for learning and regeneration?
The CNS is responsive to an ever-changing environment. Until recently, studies of neural plasticity focused almost exclusively on functional and structural changes of neuronal synapses. In recent years, myelin plasticity has emerged as a potential modulator of neural networks. Myelination of previously unmyelinated axons, and changes in the structure on already-myelinated axons, can have large effects on network function. The heterogeneity of the extent of how axons in the CNS are myelinated offers diverse scope for dynamic myelin changes to fine-tune neural circuits. The traditionally held view of myelin as a passive insulator of axons is now changing to one of lifelong changes in myelin, modulated by neuronal activity and experience. Myelin, produced by oligodendrocytes (OLs), is essential for normal brain function, as it provides fast signal transmission, promotes synchronization of neuronal signals and helps to maintain neuronal function. OLs differentiate from oligodendrocyte precursor cells (OPCs), which are distributed throughout the adult brain, and myelination continues into late adulthood. OPCs can sense neuronal activity as they receive synaptic inputs from neurons and express voltage-gated ion channels and neurotransmitter receptors, and differentiate into myelinating OLs in response to changes in neuronal activity. This lecture will explore to what extent myelin plasticity occurs in adult animals, whether myelin changes occur in non-motor learning tasks, especially in learning and memory, and questions whether myelin plasticity and myelin regeneration are two sides of the same coin.
Visualizing the multi-scale complexity of the brain
The brain is complex over multiple length-scales, from many protein molecules forming intricate nano-machines in a synapse to many neurons forming interconnected networks across the brain. Unraveling this multi-scale complexity is fundamental to our understanding of brain function and disease. In this lecture, I will introduce advances in visualizing the complex, multi-scale structures in the brain. Emphasis will be on new imaging techniques, including cryo electron tomography and correlative light-electron microscopy that enabled revealing in situ organization of synaptic molecules, and ultra-high speed volumetric imaging method VISoR developed to map brain-wide circuits at subcellular resolution. I will also discuss challenges and opportunities for interdisciplinary research collaboration to analyze and understand the enormous data generated by these cutting-edge technologies.
Creating and controlling visual environments using BonVision
Real-time rendering of closed-loop visual environments is important for next-generation understanding of brain function and behaviour, but is often prohibitively difficult for non-experts to implement and is limited to few laboratories worldwide. We developed BonVision as an easy-to-use open-source software for the display of virtual or augmented reality, as well as standard visual stimuli. BonVision has been tested on humans and mice, and is capable of supporting new experimental designs in other animal models of vision. As the architecture is based on the open-source Bonsai graphical programming language, BonVision benefits from native integration with experimental hardware. BonVision therefore enables easy implementation of closed-loop experiments, including real-time interaction with deep neural networks, and communication with behavioural and physiological measurement and manipulation devices.
Spatio-temporal large-scale organization of the trimodal connectome derived from concurrent EEG-fMRI and diffusion MRI
While time-averaged dynamics of brain functional connectivity are known to reflect the underlying structural connections, the exact relationship between large-scale function and structure remains an unsolved issue in network neuroscience. Large-scale networks are traditionally observed by correlation of fMRI timecourses, and connectivity of source-reconstructed electrophysiological measures are less prominent. Accessing the brain by using multimodal recordings combining EEG, fMRI and diffusion MRI (dMRI) can help to refine the understanding of the spatio-temporal organization of both static and dynamic brain connectivity. In this talk I will discuss our prior findings that whole-brain connectivity derived from source-reconstructed resting-state (rs) EEG is both linked to the rs-fMRI and dMRI connectome. The EEG connectome provides complimentary information to link function to structure as compared to an fMRI-only perspective. I will present an approach extending the multimodal data integration of concurrent rs-EEG-fMRI to the temporal domain by combining dynamic functional connectivity of both modalities to better understand the neural basis of functional connectivity dynamics. The close relationship between time-varying changes in EEG and fMRI whole-brain connectivity patterns provide evidence for spontaneous reconfigurations of the brain’s functional processing architecture. Finally, I will talk about data quality of connectivity derived from concurrent EEG-fMRI recordings and how the presented multimodal framework could be applied to better understand focal epilepsy. In summary this talk will give an overview of how to integrate large-scale EEG networks with MRI-derived brain structure and function. In conclusion EEG-based connectivity measures not only are closely linked to MRI-based measures of brain structure and function over different time-scales, but also provides complimentary information on the function of underlying brain organization.
Active sleep in flies: the dawn of consciousness
The brain is a prediction machine. Yet the world is never entirely predictable, for any animal. Unexpected events are surprising and this typically evokes prediction error signatures in animal brains. In humans such mismatched expectations are often associated with an emotional response as well. Appropriate emotional responses are understood to be important for memory consolidation, suggesting that valence cues more generally constitute an ancient mechanism designed to potently refine and generalize internal models of the world and thereby minimize prediction errors. On the other hand, abolishing error detection and surprise entirely is probably also maladaptive, as this might undermine the very mechanism that brains use to become better prediction machines. This paradoxical view of brain functions as an ongoing tug-of-war between prediction and surprise suggests a compelling new way to study and understand the evolution of consciousness in animals. I will present approaches to studying attention and prediction in the tiny brain of the fruit fly, Drosophila melanogaster. I will discuss how an ‘active’ sleep stage (termed rapid eye movement – REM – sleep in mammals) may have evolved in the first animal brains as a mechanism for optimizing prediction in motile creatures confronted with constantly changing environments. A role for REM sleep in emotional regulation could thus be better understood as an ancient sleep function that evolved alongside selective attention to maintain an adaptive balance between prediction and surprise. This view of active sleep has some interesting implications for the evolution of subjective awareness and consciousness.
Microbiome and behaviour: Exploring underlying mechanisms
Environmental insults alter brain function and behaviour inboth rodents and people. One putative underlying mechanism that has receivedsubstantial attention recently is the gut microbiota, the ecosystem ofsymbiotic microorganisms that populate the intestinal tract, which is known toplay a role in brain health and function via the gut-brain axis. Two keyenvironmental insults known to affect both brain function and behaviour, andthe gut microbiome, are poor diet and psychological stress. While there isstrong evidence for interactions between the microbiome and host physiology inthe context of chronic stress, little is known about the role of the microbiomein the host response to acute stress. Determining the underlying mechanisms bywhich stress may provoke functional changes in the gut and brain is criticalfor developing therapeutics to alleviate adverse consequences of traumaticstress.
Multi-scale synaptic analysis for psychiatric/emotional disorders
Dysregulation of emotional processing and its integration with cognitive functions are central features of many mental/emotional disorders associated both with externalizing problems (aggressive, antisocial behaviors) and internalizing problems (anxiety, depression). As Dr. Joseph LeDoux, our invited speaker of this program, wrote in his famous book “Synaptic self: How Our Brains Become Who We Are”—the brain’s synapses—are the channels through which we think, act, imagine, feel, and remember. Synapses encode the essence of personality, enabling each of us to function as a distinctive, integrated individual from moment to moment. Thus, exploring the functioning of synapses leads to the understanding of the mechanism of (patho)physiological function of our brain. In this context, we have investigated the pathophysiology of psychiatric disorders, with particular emphasis on the synaptic function of model mice of various psychiatric disorders such as schizophrenia, autism, depression, and PTSD. Our current interest is how synaptic inputs are integrated to generate the action potential. Because the spatiotemporal organization of neuronal firing is crucial for information processing, but how thousands of inputs to the dendritic spines drive the firing remains a central question in neuroscience. We identified a distinct pattern of synaptic integration in the disease-related models, in which extra-large (XL) spines generate NMDA spikes within these spines, which was sufficient to drive neuronal firing. We experimentally and theoretically observed that XL spines negatively correlated with working memory. Our work offers a whole new concept for dendritic computation and network dynamics, and the understanding of psychiatric research will be greatly reconsidered. The second half of my talk is the development of a novel synaptic tool. Because, no matter how beautifully we can illuminate the spine morphology and how accurately we can quantify the synaptic integration, the links between synapse and brain function remain correlational. In order to challenge the causal relationship between synapse and brain function, we established AS-PaRac1, which is unique not only because it can specifically label and manipulate the recently potentiated dendritic spine (Hayashi-Takagi et al, 2015, Nature). With use of AS-PaRac1, we developed an activity-dependent simultaneous labeling of the presynaptic bouton and the potentiated spines to establish “functional connectomics” in a synaptic resolution. When we apply this new imaging method for PTSD model mice, we identified a completely new functional neural circuit of brain region A→B→C with a very strong S/N in the PTSD model mice. This novel tool of “functional connectomics” and its photo-manipulation could open up new areas of emotional/psychiatric research, and by extension, shed light on the neural networks that determine who we are.
Making memories in mice
Understanding how the brain uses information is a fundamental goal of neuroscience. Several human disorders (ranging from autism spectrum disorder to PTSD to Alzheimer’s disease) may stem from disrupted information processing. Therefore, this basic knowledge is not only critical for understanding normal brain function, but also vital for the development of new treatment strategies for these disorders. Memory may be defined as the retention over time of internal representations gained through experience, and the capacity to reconstruct these representations at later times. Long-lasting physical brain changes (‘engrams’) are thought to encode these internal representations. The concept of a physical memory trace likely originated in ancient Greece, although it wasn’t until 1904 that Richard Semon first coined the term ‘engram’. Despite its long history, finding a specific engram has been challenging, likely because an engram is encoded at multiple levels (epigenetic, synaptic, cell assembly). My lab is interested in understanding how specific neurons are recruited or allocated to an engram, and how neuronal membership in an engram may change over time or with new experience. Here I will describe both older and new unpublished data in our efforts to understand memories in mice.
Towards a neurally mechanistic understanding of visual cognition
I am interested in developing a neurally mechanistic understanding of how primate brains represent the world through its visual system and how such representations enable a remarkable set of intelligent behaviors. In this talk, I will primarily highlight aspects of my current research that focuses on dissecting the brain circuits that support core object recognition behavior (primates’ ability to categorize objects within hundreds of milliseconds) in non-human primates. On the one hand, my work empirically examines how well computational models of the primate ventral visual pathways embed knowledge of the visual brain function (e.g., Bashivan*, Kar*, DiCarlo, Science, 2019). On the other hand, my work has led to various functional and architectural insights that help improve such brain models. For instance, we have exposed the necessity of recurrent computations in primate core object recognition (Kar et al., Nature Neuroscience, 2019), one that is strikingly missing from most feedforward artificial neural network models. Specifically, we have observed that the primate ventral stream requires fast recurrent processing via ventrolateral PFC for robust core object recognition (Kar and DiCarlo, Neuron, 2021). In addition, I have been currently developing various chemogenetic strategies to causally target specific bidirectional neural circuits in the macaque brain during multiple object recognition tasks to further probe their relevance during this behavior. I plan to transform these data and insights into tangible progress in neuroscience via my collaboration with various computational groups and building improved brain models of object recognition. I hope to end the talk with a brief glimpse of some of my planned future work!
Generative models of the human connectome
The human brain is a complex network of neuronal connections. The precise arrangement of these connections, otherwise known as the topology of the network, is crucial to its functioning. Recent efforts to understand how the complex topology of the brain has emerged have used generative mathematical models, which grow synthetic networks according to specific wiring rules. Evidence suggests that a wiring rule which emulates a trade-off between connection costs and functional benefits can produce networks that capture essential topological properties of brain networks. In this webinar, Professor Alex Fornito and Dr Stuart Oldham will discuss these previous findings, as well as their own efforts in creating more physiologically constrained generative models. Professor Alex Fornito is Head of the Brain Mapping and Modelling Research Program at the Turner Institute for Brain and Mental Health. His research focuses on developing new imaging techniques for mapping human brain connectivity and applying these methods to shed light on brain function in health and disease. Dr Stuart Oldham is a Research Fellow at the Turner Institute for Brain and Mental Health and a Research Officer at the Murdoch Children’s Research Institute. He is interested in characterising the organisation of human brain networks, with particular focus on how this organisation develops, using neuroimaging and computational tools.
Prof. Humphries reads from "The Spike" 📖
We see the last cookie in the box and think, can I take that? We reach a hand out. In the 2.1 seconds that this impulse travels through our brain, billions of neurons communicate with one another, sending blips of voltage through our sensory and motor regions. Neuroscientists call these blips “spikes.” Spikes enable us to do everything: talk, eat, run, see, plan, and decide. In The Spike, Mark Humphries takes readers on the epic journey of a spike through a single, brief reaction. In vivid language, Humphries tells the story of what happens in our brain, what we know about spikes, and what we still have left to understand about them. Drawing on decades of research in neuroscience, Humphries explores how spikes are born, how they are transmitted, and how they lead us to action. He dives into previously unanswered mysteries: Why are most neurons silent? What causes neurons to fire spikes spontaneously, without input from other neurons or the outside world? Why do most spikes fail to reach any destination? Humphries presents a new vision of the brain, one where fundamental computations are carried out by spontaneous spikes that predict what will happen in the world, helping us to perceive, decide, and react quickly enough for our survival. Traversing neuroscience’s expansive terrain, The Spike follows a single electrical response to illuminate how our extraordinary brains work.
Mathematical models of neurodegenerative diseases
Neurodegenerative diseases such as Alzheimer’s or Parkinson’s are devastating conditions with poorly understood mechanisms and no cure. Yet, a striking feature of these conditions is the characteristic pattern of invasion throughout the brain, leading to well-codified disease stages associated with various cognitive deficits and pathologies. How can we use mathematical modelling to gain insight into this process and, doing so, gain understanding about how the brain works? In this talk, I will show that by linking new mathematical theories to recent progress in imaging, we can unravel some of the universal features associated with dementia and, more generally, brain functions.
From 1D to 5D: Data-driven Discovery of Whole-brain Dynamic Connectivity in fMRI Data
The analysis of functional magnetic resonance imaging (fMRI) data can greatly benefit from flexible analytic approaches. In particular, the advent of data-driven approaches to identify whole-brain time-varying connectivity and activity has revealed a number of interesting relevant variation in the data which, when ignored, can provide misleading information. In this lecture I will provide a comparative introduction of a range of data-driven approaches to estimating time-varying connectivity. I will also present detailed examples where studies of both brain health and disorder have been advanced by approaches designed to capture and estimate time-varying information in resting fMRI data. I will review several exemplar data sets analyzed in different ways to demonstrate the complementarity as well as trade-offs of various modeling approaches to answer questions about brain function. Finally, I will review and provide examples of strategies for validating time-varying connectivity including simulations, multimodal imaging, and comparative prediction within clinical populations, among others. As part of the interactive aspect I will provide a hands-on guide to the dynamic functional network connectivity toolbox within the GIFT software, including an online didactic analytic decision tree to introduce the various concepts and decisions that need to be made when using such tools
BrainGlobe: a Python ecosystem for computational (neuro)anatomy
Neuroscientists routinely perform experiments aimed at recording or manipulating neural activity, uncovering physiological processes underlying brain function or elucidating aspects of brain anatomy. Understanding how the brain generates behaviour ultimately depends on merging the results of these experiments into a unified picture of brain anatomy and function. We present BrainGlobe, a new initiative aimed at developing common Python tools for computational neuroanatomy. These include cellfinder for fast, accurate cell detection in whole-brain microscopy images, brainreg for aligning images to a reference atlas, and brainrender for visualisation of anatomically registered data. These software packages are developed around the BrainGlobe Atlas API. This API provides a common Python interface to download and interact with reference brain atlases from multiple species (including human, mouse and larval zebrafish). This allows software to be developed agnostic to the atlas and species, increasing adoption and interoperability of software tools in neuroscience.
Workshop: Spatial Brain Dynamics
Traditionally, the term dynamics means changes in a system evolving over time. However, in the brain action potentials propagate along axons to induce postsynaptic currents with different delays at many sites simultaneously. This fundamental computational mechanism evolves spatially to engage the neuron populations involved in brain functions. To identify and understand the spatial processing in brains, this workshop will focus on the spatial principles of brain dynamics that determine how action potentials and membrane currents propagate in the networks of neurons that brains are made of. We will focus on non-artificial dynamics, which excludes in vitro dynamics, interference, electrical and optogenetic stimulations of brains in vivo. Recent non-artificial studies of spatial brain dynamics can actually explain how sensory, motor and internal brain functions evolve. The purpose of this workshop is to discuss these recent results and identify common principles of spatial brain dynamics.
Fragility of the human connectome across the lifespan
The human brain network architecture can reveal crucial aspects of brain function and dysfunction. The topology of this network (known as the connectome) is shaped by a trade-off between wiring cost and network efficiency, and it has highly connected hub regions playing a prominent role in many brain disorders. By studying a landscape of plausible brain networks that preserve the wiring cost, fragile and resilient hubs can be identified. In this webinar, Dr Leonardo Gollo and Dr James Pang from Monash University will discuss this approach across the lifespan and some of its implications for neurodevelopmental and neurodegenerative diseases. Dr Leonardo Gollo is a Senior Research Fellow at the Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University. He holds an ARC Future Fellowship and his research interests include brain modelling, systems neuroscience, and connectomics. Dr James Pang is a Research Fellow at the Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University. His research interests are on combining neuroimaging and biophysical modelling to better understand the mechanisms of brain function in health and disease.
Workshop: Spatial Brain Dynamics
Traditionally, the term dynamics means changes in a system evolving over time. However, in the brain action potentials propagate along axons to induce postsynaptic currents with different delays at many sites simultaneously. This fundamental computational mechanism evolves spatially to engage the neuron populations involved in brain functions. To identify and understand the spatial processing in brains, this workshop will focus on the spatial principles of brain dynamics that determine how action potentials and membrane currents propagate in the networks of neurons that brains are made of. We will focus on non-artificial dynamics, which excludes in vitro dynamics, interference, electrical and optogenetic stimulations of brains in vivo. Recent non-artificial studies of spatial brain dynamics can actually explain how sensory, motor and internal brain functions evolve. The purpose of this workshop is to discuss these recent results and identify common principles of spatial brain dynamics.
Workshop: Spatial Brain Dynamics
Traditionally, the term dynamics means changes in a system evolving over time. However, in the brain action potentials propagate along axons to induce postsynaptic currents with different delays at many sites simultaneously. This fundamental computational mechanism evolves spatially to engage the neuron populations involved in brain functions. To identify and understand the spatial processing in brains, this workshop will focus on the spatial principles of brain dynamics that determine how action potentials and membrane currents propagate in the networks of neurons that brains are made of. We will focus on non-artificial dynamics, which excludes in vitro dynamics, interference, electrical and optogenetic stimulations of brains in vivo. Recent non-artificial studies of spatial brain dynamics can actually explain how sensory, motor and internal brain functions evolve. The purpose of this workshop is to discuss these recent results and identify common principles of spatial brain dynamics.
How dendrites help solve biological and machine learning problems
Dendrites are thin processes that extend from the cell body of neurons, the main computing units of the brain. The role of dendrites in complex brain functions has been investigated for several decades, yet their direct involvement in key behaviors such as for example sensory perception has only recently been established. In my presentation I will discuss how computational modelling has helped us illuminate dendritic function. I will present the main findings of a number of projects in lab dealing with dendritic nonlinearities in excitatory and inhibitory and their consequences on neuronal tuning and memory formation, the role of dendrites in solving nonlinear problems in human neurons and recent efforts to advance machine learning algorithms by adopting dendritic features.
Artificial neural networks do not adequately mimic whatever is going on in the real brain
One may think that Deep Learning technology works in ways that are similar to the human brain. This is not really true. Our best AI technology still does not mimic the brain sufficiently well to be a match in intelligence. I will describe seven differences on how our minds work in ways diametrically opposite to those of Deep Learning technology.
Magnetic Resonance Measures of Brain Blood Vessels, Metabolic Activity, and Pathology in Multiple Sclerosis
The normally functioning blood-brain barrier (BBB) regulates the transfer of material between blood and brain. BBB dysfunction has long been recognized in multiple sclerosis (MS), and there is considerable interest in quantifying functional aspects of brain blood vessels and their role in disease progression. Parenchymal water content and its association with volume regulation is important for proper brain function, and is one of the key roles of the BBB. There is convincing evidence that the astrocyte is critical in establishing and maintaining a functional BBB and providing metabolic support to neurons. Increasing evidence suggests that functional interactions between endothelia, pericytes, astrocytes, and neurons, collectively known as the neurovascular unit, contribute to brain water regulation, capillary blood volume and flow, BBB permeability, and are responsive to metabolic demands. Increasing evidence suggests altered metabolism in MS brain which may contribute to reduced neuro-repair and increased neurodegeneration. Metabolically relevant biomarkers may provide sensitive readouts of brain tissue at risk of degeneration, and magnetic resonance offers substantial promise in this regard. Dynamic contrast enhanced MRI combined with appropriate pharmacokinetic modeling allows quantification of distinct features of BBB including permeabilities to contrast agent and water, with rate constants that differ by six orders of magnitude. Mapping of these rate constants provides unique biological aspects of brain vasculature relevant to MS.
New Strategies and Approaches to Tackle and Understand Neurological Disorder
Broadly, the Mauro Costa-Mattioli laboratory (The MCM Lab) encompasses two complementary lines of research. The first one, more traditional but very important, aims at unraveling the molecular mechanisms underlying memory formation (e.g., using state-of-the-art molecular and cell-specific genetic approaches). Learning and memory disorders can strike the brain during development (e.g., Autism Spectrum Disorders and Down Syndrome), as well as during adulthood (e.g., Alzheimer’s disease). We are interested in understanding the specific circuits and molecular pathways that are primarily targeted in these disorders and how they can be restored. To tackle these questions, we use a multidisciplinary, convergent and cross-species approach that combines mouse and fly genetics, molecular biology, electrophysiology, stem cell biology, optogenetics and behavioral techniques. The second line of research, more recent and relatively unexplored, is focused on understanding how gut microbes control CNS driven-behavior and brain function. Our recent discoveries, that microbes in the gut could modulate brain function and behavior in a very powerful way, have added a whole new dimension to the classic view of how complex behaviors are controlled. The unexpected findings have opened new avenues of study for us and are currently driving my lab to answer a host of new and very interesting questions: - What are the gut microbes (and metabolites) that regulate CNS-driven behaviors? Would it be possible to develop an unbiased screening method to identify specific microbes that regulate different behaviors? - If this is the case, can we identify how members of the gut microbiome (and their metabolites) mechanistically influence brain function? - What is the communication channel between the gut microbiota and the brain? Do different gut microbes use different ways to interact with the brain? - Could disruption of the gut microbial ecology cause neurodevelopmental dysfunction? If so, what is the impact of disruption in young and adult animals? - More importantly, could specific restoration of selected bacterial strains (new generation probiotics) represent a novel therapeutic approach for the targeted treatment of neurodevelopmental disorders? - Finally, can we develop microbiota-directed therapeutic foods to repair brain dysfunction in a variety of neurological disorders?
How the immune system shapes synaptic functions
The synapse is the core component of the nervous system and synapse formation is the critical step in the assembly of neuronal circuits. The assembly and maturation of synapses requires the contribution of secreted and membrane-associated proteins, with neuronal activity playing crucial roles in regulating synaptic strength, neuronal membrane properties, and neural circuit refinement. The molecular mechanisms of synapse assembly and refinement have been so far largely examined on a gene-by-gene basis and with a perspective fully centered on neuronal cells. However, in the last years, the involvement of non-neuronal cells has emerged. Among these, microglia, the resident immune cells of the central nervous system, have been shown to play a key role in synapse formation and elimination. Contacts of microglia with dendrites in the somatosensory cortex were found to induce filopodia and dendritic spines via Ca2+ and actin-dependent processes, while microglia-derived BDNF was shown to promote learning-dependent synapse formation. Microglia is also recognized to have a central role in the widespread elimination (or pruning) of exuberant synaptic connections during development. Clarifying the processes by which microglia control synapse homeostasis is essential to advance our current understanding of brain functions. Clear answers to these questions will have important implications for our understanding of brain diseases, as the fact that many psychiatric and neurological disorders are synaptopathies (i.e. diseases of the synapse) is now widely recognized. In the last years, my group has identified TREM2, an innate immune receptor with phagocytic and antiinflammatory properties expressed in brain exclusively by microglia, as essential for microglia-mediated synaptic refinement during the early stages of brain development. The talk will describe the role of TREM2 in synapse elimination and introduce the molecular actors involved. I will also describe additional pathways by which the immune system may affect the formation and homeostasis of synaptic contacts.
Markers of brain connectivity and sleep-dependent restoration: basic research and translation into clinical populations
The human brain is a heavily interconnected structure giving rise to complex functions. While brain functionality is mostly revealed during wakefulness, the sleeping brain might offer another view into physiological and pathological brain connectivity. Furthermore, there is a large body of evidence supporting that sleep mediates plastic changes in brain connectivity. Although brain plasticity depends on environmental input which is provided in the waking state, disconnection during sleep might be necessary for integrating new into existing information and at the same time restoring brain efficiency. In this talk, I will present structural, molecular, and electrophysiological markers of brain connectivity and sleep-dependent restoration that we have evaluated using Magnetic Resonance Imaging and electroencephalography in a healthy population. In a second step, I will show how we translated the gained findings into two clinical populations in which alterations in brain connectivity have been described, the neuropsychiatric disorder attention-deficit/hyperactivity disorder (ADHD) and the neurologic disorder thalamic ischemic stroke.
Mapping early brain network changes in neurodegenerative and cerebrovascular disorders: a longitudinal perspective
The spatial patterning of each neurodegenerative disease relates closely to a distinct structural and functional network in the human brain. This talk will mainly describe how brain network-sensitive neuroimaging methods such as resting-state fMRI and diffusion MRI can shed light on brain network dysfunctions associated with pathology and cognitive decline from preclinical to clinical dementia. I will first present our findings from two independent datasets on how amyloid and cerebrovascular pathology influence brain functional networks cross-sectionally and longitudinally in individuals with mild cognitive impairment and dementia. Evidence on longitudinal functional network organizational changes in healthy older adults and the influence of APOE genotype will be presented. In the second part, I will describe our work on how different pathology influences brain structural network and white matter microstructure. I will also touch on some new data on how brain network integrity contributes to behavior and disease progression using multivariate or machine learning approaches. These findings underscore the importance of studying selective brain network vulnerability instead of individual region and longitudinal design. Further developed with machine learning approaches, multimodal network-specific imaging signatures will help reveal disease mechanisms and facilitate early detection, prognosis and treatment search of neuropsychiatric disorders.
Direct measurement of whole-brain functional connectivity in C. elegans
COSYNE 2022
Deciphering brain function through in vivo simultaneous multi-region neuronal level brain imaging of freely behaving animals
FENS Forum 2024
Development of the whole-brain functional connectome explored via graph theory analysis
FENS Forum 2024
The impact of high-fat diet on microglial cells and social behavior in mice: Implications for diet-induced changes in brain function
FENS Forum 2024
Modeling and inference of brain functional connectivity networks in the Shank2 mouse model of autism using functional ultrasound
FENS Forum 2024
Multivariate pattern analysis on associations between resting-state whole-brain functional connectivity patterns and medial prefrontal GABA levels specific to major depressive disorder
FENS Forum 2024
Oblique light sheet tracking microscopy for whole brain functional imaging in a freely swimming zebrafish larva
FENS Forum 2024
Screening AAV delivery routes, capsids, and promoters for cortex-wide functional and long-term stable access to brain function in large-brain species
FENS Forum 2024
Social drinking: Reward-related brain function and daily life correlates
FENS Forum 2024
Unlocking the role of Enhancer of Polycomb Homolog 1 (EPC1) in brain function
FENS Forum 2024