human brain
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
Cellular Crosstalk in Brain Development, Evolution and Disease
Cellular crosstalk is an essential process during brain development and is influenced by numerous factors, including cell morphology, adhesion, the local extracellular matrix and secreted vesicles. Inspired by mutations associated with neurodevelopmental disorders, we focus on understanding the role of extracellular mechanisms essential for the proper development of the human brain. Therefore, we combine 2D and 3D in vitro human models to better understand the molecular and cellular mechanisms involved in progenitor proliferation and fate, migration and maturation of excitatory and inhibitory neurons during human brain development and tackle the causes of neurodevelopmental disorders.
Functional Imaging of the Human Brain: A Window into the Organization of the Human Mind
Expanding mechanisms and therapeutic targets for neurodegenerative disease
A hallmark pathological feature of the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is the depletion of RNA-binding protein TDP-43 from the nucleus of neurons in the brain and spinal cord. A major function of TDP-43 is as a repressor of cryptic exon inclusion during RNA splicing. By re-analyzing RNA-sequencing datasets from human FTD/ALS brains, we discovered dozens of novel cryptic splicing events in important neuronal genes. Single nucleotide polymorphisms in UNC13A are among the strongest hits associated with FTD and ALS in human genome-wide association studies, but how those variants increase risk for disease is unknown. We discovered that TDP-43 represses a cryptic exon-splicing event in UNC13A. Loss of TDP-43 from the nucleus in human brain, neuronal cell lines and motor neurons derived from induced pluripotent stem cells resulted in the inclusion of a cryptic exon in UNC13A mRNA and reduced UNC13A protein expression. The top variants associated with FTD or ALS risk in humans are located in the intron harboring the cryptic exon, and we show that they increase UNC13A cryptic exon splicing in the face of TDP-43 dysfunction. Together, our data provide a direct functional link between one of the strongest genetic risk factors for FTD and ALS (UNC13A genetic variants), and loss of TDP-43 function. Recent analyses have revealed even further changes in TDP-43 target genes, including widespread changes in alternative polyadenylation, impacting expression of disease-relevant genes (e.g., ELP1, NEFL, and TMEM106B) and providing evidence that alternative polyadenylation is a new facet of TDP-43 pathology.
Functional Plasticity in the Language Network – evidence from Neuroimaging and Neurostimulation
Efficient cognition requires flexible interactions between distributed neural networks in the human brain. These networks adapt to challenges by flexibly recruiting different regions and connections. In this talk, I will discuss how we study functional network plasticity and reorganization with combined neurostimulation and neuroimaging across the adult life span. I will argue that short-term plasticity enables flexible adaptation to challenges, via functional reorganization. My key hypothesis is that disruption of higher-level cognitive functions such as language can be compensated for by the recruitment of domain-general networks in our brain. Examples from healthy young brains illustrate how neurostimulation can be used to temporarily interfere with efficient processing, probing short-term network plasticity at the systems level. Examples from people with dyslexia help to better understand network disorders in the language domain and outline the potential of facilitatory neurostimulation for treatment. I will also discuss examples from aging brains where plasticity helps to compensate for loss of function. Finally, examples from lesioned brains after stroke provide insight into the brain’s potential for long-term reorganization and recovery of function. Collectively, these results challenge the view of a modular organization of the human brain and argue for a flexible redistribution of function via systems plasticity.
Cognitive maps, navigational strategies, and the human brain
Learning Representations of Complex Meaning in the Human Brain
Reactivation in the human brain connects the past with the present
Spatial Organization of Cellular Reactive States in Human Brain Cancer
Applied cognitive neuroscience to improve learning and therapeutics
Advancements in cognitive neuroscience have provided profound insights into the workings of the human brain and the methods used offer opportunities to enhance performance, cognition, and mental health. Drawing upon interdisciplinary collaborations in the University of California San Diego, Human Performance Optimization Lab, this talk explores the application of cognitive neuroscience principles in three domains to improve human performance and alleviate mental health challenges. The first section will discuss studies addressing the role of vision and oculomotor function in athletic performance and the potential to train these foundational abilities to improve performance and sports outcomes. The second domain considers the use of electrophysiological measurements of the brain and heart to detect, and possibly predict, errors in manual performance, as shown in a series of studies with surgeons as they perform robot-assisted surgery. Lastly, findings from clinical trials testing personalized interventional treatments for mood disorders will be discussed in which the temporal and spatial parameters of transcranial magnetic stimulation (TMS) are individualized to test if personalization improves treatment response and can be used as predictive biomarkers to guide treatment selection. Together, these translational studies use the measurement tools and constructs of cognitive neuroscience to improve human performance and well-being.
Characterizing the causal role of large-scale network interactions in supporting complex cognition
Neuroimaging has greatly extended our capacity to study the workings of the human brain. Despite the wealth of knowledge this tool has generated however, there are still critical gaps in our understanding. While tremendous progress has been made in mapping areas of the brain that are specialized for particular stimuli, or cognitive processes, we still know very little about how large-scale interactions between different cortical networks facilitate the integration of information and the execution of complex tasks. Yet even the simplest behavioral tasks are complex, requiring integration over multiple cognitive domains. Our knowledge falls short not only in understanding how this integration takes place, but also in what drives the profound variation in behavior that can be observed on almost every task, even within the typically developing (TD) population. The search for the neural underpinnings of individual differences is important not only philosophically, but also in the service of precision medicine. We approach these questions using a three-pronged approach. First, we create a battery of behavioral tasks from which we can calculate objective measures for different aspects of the behaviors of interest, with sufficient variance across the TD population. Second, using these individual differences in behavior, we identify the neural variance which explains the behavioral variance at the network level. Finally, using covert neurofeedback, we perturb the networks hypothesized to correspond to each of these components, thus directly testing their casual contribution. I will discuss our overall approach, as well as a few of the new directions we are currently pursuing.
Modeling human brain development and disease: the role of primary cilia
Neurodevelopmental disorders (NDDs) impose a global burden, affecting an increasing number of individuals. While some causative genes have been identified, understanding the human-specific mechanisms involved in these disorders remains limited. Traditional gene-driven approaches for modeling brain diseases have failed to capture the diverse and convergent mechanisms at play. Centrosomes and cilia act as intermediaries between environmental and intrinsic signals, regulating cellular behavior. Mutations or dosage variations disrupting their function have been linked to brain formation deficits, highlighting their importance, yet their precise contributions remain largely unknown. Hence, we aim to investigate whether the centrosome/cilia axis is crucial for brain development and serves as a hub for human-specific mechanisms disrupted in NDDs. Towards this direction, we first demonstrated species-specific and cell-type-specific differences in the cilia-genes expression during mouse and human corticogenesis. Then, to dissect their role, we provoked their ectopic overexpression or silencing in the developing mouse cortex or in human brain organoids. Our findings suggest that cilia genes manipulation alters both the numbers and the position of NPCs and neurons in the developing cortex. Interestingly, primary cilium morphology is disrupted, as we find changes in their length, orientation and number that lead to disruption of the apical belt and altered delamination profiles during development. Our results give insight into the role of primary cilia in human cortical development and address fundamental questions regarding the diversity and convergence of gene function in development and disease manifestation. It has the potential to uncover novel pharmacological targets, facilitate personalized medicine, and improve the lives of individuals affected by NDDs through targeted cilia-based therapies.
Contrasting developmental principles of human brain development and their relevance to neurodevelopmental disorders
Mitochondrial diversity in the mouse and human brain
The basis of the mind, of mental states, and complex behaviors is the flow of energy through microscopic and macroscopic brain structures. Energy flow through brain circuits is powered by thousands of mitochondria populating the inside of every neuron, glial, and other nucleated cell across the brain-body unit. This seminar will cover emerging approaches to study the mind-mitochondria connection and present early attempts to map the distribution and diversity of mitochondria across brain tissue. In rodents, I will present convergent multimodal evidence anchored in enzyme activities, gene expression, and animal behavior that distinct behaviorally-relevant mitochondrial phenotypes exist across large-scale mouse brain networks. Extending these findings to the human brain, I will present a developing systematic biochemical and molecular map of mitochondrial variation across cortical and subcortical brain structures, representing a foundation to understand the origin of complex energy patterns that give rise to the human mind.
Executive functions in the brain of deaf individuals – sensory and language effects
Executive functions are cognitive processes that allow us to plan, monitor and execute our goals. Using fMRI, we investigated how early deafness influences crossmodal plasticity and the organisation of executive functions in the adult human brain. Results from a range of visual executive function tasks (working memory, task switching, planning, inhibition) show that deaf individuals specifically recruit superior temporal “auditory” regions during task switching. Neural activity in auditory regions predicts behavioural performance during task switching in deaf individuals, highlighting the functional relevance of the observed cortical reorganisation. Furthermore, language grammatical skills were correlated with the level of activation and functional connectivity of fronto-parietal networks. Together, these findings show the interplay between sensory and language experience in the organisation of executive processing in the brain.
The quest for brain identification
In the 17th century, physician Marcello Malpighi observed the existence of distinctive patterns of ridges and sweat glands on fingertips. This was a major breakthrough, and originated a long and continuing quest for ways to uniquely identify individuals based on fingerprints, a technique massively used until today. It is only in the past few years that technologies and methodologies have achieved high-quality measures of an individual’s brain to the extent that personality traits and behavior can be characterized. The concept of “fingerprints of the brain” is very novel and has been boosted thanks to a seminal publication by Finn et al. in 2015. They were among the firsts to show that an individual’s functional brain connectivity profile is both unique and reliable, similarly to a fingerprint, and that it is possible to identify an individual among a large group of subjects solely on the basis of her or his connectivity profile. Yet, the discovery of brain fingerprints opened up a plethora of new questions. In particular, what exactly is the information encoded in brain connectivity patterns that ultimately leads to correctly differentiating someone’s connectome from anybody else’s? In other words, what makes our brains unique? In this talk I am going to partially address these open questions while keeping a personal viewpoint on the subject. I will outline the main findings, discuss potential issues, and propose future directions in the quest for identifiability of human brain networks.
Cellular and genetic mechanisms of cerebral cortex folding
One of the most prominent features of the human brain is the fabulous size of the cerebral cortex and its intricate folding, both of which emerge during development. Over the last few years, work from my lab has shown that specific cellular and genetic mechanisms play central roles in cortex folding, particularly linked to neural stem and progenitor cells. Key mechanisms include high rates of neurogenesis, high abundance of basal Radial Glia Cells (bRGCs), and neuron migration, all of which are intertwined during development. We have also shown that primary cortical folds follow highly stereotyped patterns, defined by a spatial-temporal protomap of gene expression within germinal layers of the developing cortex. I will present recent findings from my laboratory revealing novel cellular and genetic mechanisms that regulate cortex expansion and folding. We have uncovered the contribution of epigenetic regulation to the establishment of the cortex folding protomap, modulating the expression levels of key transcription factors that control progenitor cell proliferation and cortex folding. At the single cell level, we have identified an unprecedented diversity of cortical progenitor cell classes in the ferret and human embryonic cortex. These are differentially enriched in gyrus versus sulcus regions and establish parallel cell lineages, not observed in mouse. Our findings show that genetic and epigenetic mechanisms in gyrencephalic species diversify cortical progenitor cell types and implement parallel cell linages, driving the expansion of neurogenesis and patterning cerebral cortex folds.
Trends in NeuroAI - Meta's MEG-to-image reconstruction
Trends in NeuroAI is a reading group hosted by the MedARC Neuroimaging & AI lab (https://medarc.ai/fmri). Title: Brain-optimized inference improves reconstructions of fMRI brain activity Abstract: The release of large datasets and developments in AI have led to dramatic improvements in decoding methods that reconstruct seen images from human brain activity. We evaluate the prospect of further improving recent decoding methods by optimizing for consistency between reconstructions and brain activity during inference. We sample seed reconstructions from a base decoding method, then iteratively refine these reconstructions using a brain-optimized encoding model that maps images to brain activity. At each iteration, we sample a small library of images from an image distribution (a diffusion model) conditioned on a seed reconstruction from the previous iteration. We select those that best approximate the measured brain activity when passed through our encoding model, and use these images for structural guidance during the generation of the small library in the next iteration. We reduce the stochasticity of the image distribution at each iteration, and stop when a criterion on the "width" of the image distribution is met. We show that when this process is applied to recent decoding methods, it outperforms the base decoding method as measured by human raters, a variety of image feature metrics, and alignment to brain activity. These results demonstrate that reconstruction quality can be significantly improved by explicitly aligning decoding distributions to brain activity distributions, even when the seed reconstruction is output from a state-of-the-art decoding algorithm. Interestingly, the rate of refinement varies systematically across visual cortex, with earlier visual areas generally converging more slowly and preferring narrower image distributions, relative to higher-level brain areas. Brain-optimized inference thus offers a succinct and novel method for improving reconstructions and exploring the diversity of representations across visual brain areas. Speaker: Reese Kneeland is a Ph.D. student at the University of Minnesota working in the Naselaris lab. Paper link: https://arxiv.org/abs/2312.07705
Trends in NeuroAI - Meta's MEG-to-image reconstruction
Trends in NeuroAI is a reading group hosted by the MedARC Neuroimaging & AI lab (https://medarc.ai/fmri). This will be an informal journal club presentation, we do not have an author of the paper joining us. Title: Brain decoding: toward real-time reconstruction of visual perception Abstract: In the past five years, the use of generative and foundational AI systems has greatly improved the decoding of brain activity. Visual perception, in particular, can now be decoded from functional Magnetic Resonance Imaging (fMRI) with remarkable fidelity. This neuroimaging technique, however, suffers from a limited temporal resolution (≈0.5 Hz) and thus fundamentally constrains its real-time usage. Here, we propose an alternative approach based on magnetoencephalography (MEG), a neuroimaging device capable of measuring brain activity with high temporal resolution (≈5,000 Hz). For this, we develop an MEG decoding model trained with both contrastive and regression objectives and consisting of three modules: i) pretrained embeddings obtained from the image, ii) an MEG module trained end-to-end and iii) a pretrained image generator. Our results are threefold: Firstly, our MEG decoder shows a 7X improvement of image-retrieval over classic linear decoders. Second, late brain responses to images are best decoded with DINOv2, a recent foundational image model. Third, image retrievals and generations both suggest that MEG signals primarily contain high-level visual features, whereas the same approach applied to 7T fMRI also recovers low-level features. Overall, these results provide an important step towards the decoding - in real time - of the visual processes continuously unfolding within the human brain. Speaker: Dr. Paul Scotti (Stability AI, MedARC) Paper link: https://arxiv.org/abs/2310.19812
Deep language models as a cognitive model for natural language processing in the human brain
A synergistic core for human brain evolution and cognition
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.
Neuroinflammation in Epilepsy: what have we learned from human brain tissue specimens ?
Epileptogenesis is a gradual and dynamic process leading to difficult-to-treat seizures. Several cellular, molecular, and pathophysiologic mechanisms, including the activation of inflammatory processes. The use of human brain tissue represents a crucial strategy to advance our understanding of the underlying neuropathology and the molecular and cellular basis of epilepsy and related cognitive and behavioral comorbidities, The mounting evidence obtained during the past decade has emphasized the critical role of inflammation in the pathophysiological processes implicated in a large spectrum of genetic and acquired forms of focal epilepsies. Dissecting the cellular and molecular mediators of the pathological immune responses and their convergent and divergent mechanisms, is a major requisite for delineating their role in the establishment of epileptogenic networks. The role of small regulatory molecules involved in the regulation of specific pro- and anti-inflammatory pathways and the crosstalk between neuroinflammation and oxidative stress will be addressed. The observations supporting the activation of both innate and adaptive immune responses in human focal epilepsy will be discussed and elaborated, highlighting specific inflammatory pathways as potential targets for antiepileptic, disease-modifying therapeutic strategies.
Use of brain imaging data to improve prescriptions of psychotropic drugs - Examples of ketamine in depression and antipsychotics in schizophrenia
The use of molecular imaging, particularly PET and SPECT, has significantly transformed the treatment of schizophrenia with antipsychotic drugs since the late 1980s. It has offered insights into the links between drug target engagement, clinical effects, and side effects. A therapeutic window for receptor occupancy is established for antipsychotics, yet there is a divergence of opinions regarding the importance of blood levels, with many downplaying their significance. As a result, the role of therapeutic drug monitoring (TDM) as a personalized therapy tool is often underrated. Since molecular imaging of antipsychotics has focused almost entirely on D2-like dopamine receptors and their potential to control positive symptoms, negative symptoms and cognitive deficits are hardly or not at all investigated. Alternative methods have been introduced, i.e. to investigate the correlation between approximated receptor occupancies from blood levels and cognitive measures. Within the domain of antidepressants, and specifically regarding ketamine's efficacy in depression treatment, there is limited comprehension of the association between plasma concentrations and target engagement. The measurement of AMPA receptors in the human brain has added a new level of comprehension regarding ketamine's antidepressant effects. To ensure precise prescription of psychotropic drugs, it is vital to have a nuanced understanding of how molecular and clinical effects interact. Clinician scientists are assigned with the task of integrating these indispensable pharmacological insights into practice, thereby ensuring a rational and effective approach to the treatment of mental health disorders, signaling a new era of personalized drug therapy mechanisms that promote neuronal plasticity not only under pathological conditions, but also in the healthy aging brain.
NII Methods (journal club): NeuroQuery, comprehensive meta-analysis of human brain mapping
We will discuss a recent paper by Taylor et al. (2023): https://www.sciencedirect.com/science/article/pii/S1053811923002896. They discuss the merits of highlighting results instead of hiding them; that is, clearly marking which voxels and clusters pass a given significance threshold, but still highlighting sub-threshold results, with opacity proportional to the strength of the effect. They use this to illustrate how there in fact may be more agreement between researchers than previously thought, using the NARPS dataset as an example. By adopting a continuous, "highlighted" approach, it becomes clear that the majority of effects are in the same location and that the effect size is in the same direction, compared to an approach that only permits rejecting or not rejecting the null hypothesis. We will also talk about the implications of this approach for creating figures, detecting artifacts, and aiding reproducibility.
Cellular crosstalk in Neurodevelopmental Disorders
Cellular crosstalk is an essential process during brain development and it is influenced by numerous factors, including the morphology of the cells, their adhesion molecules, the local extracellular matrix and the secreted vesicles. Inspired by mutations associated with neurodevelopmental disorders, we focus on understanding the role of extracellular mechanisms essential for the correct development of the human brain. Hence, we combine the in vivo mouse model and the in vitro human-derived neurons, cerebral organoids, and dorso-ventral assembloids in order to better comprehend the molecular and cellular mechanisms involved in ventral progenitors’ proliferation and fate as well as migration and maturation of inhibitory neurons during human brain development and tackle the causes of neurodevelopmental disorders. We particularly focus on mutations in genes influencing cell-cell contacts, extracellular matrix, and secretion of vesicles and therefore study intrinsic and extrinsic mechanisms contributing to the formation of the brain. Our data reveal an important contribution of cell non-autonomous mechanisms in the development of neurodevelopmental disorders.
NII Methods (journal club): NeuroQuery, comprehensive meta-analysis of human brain mapping
We will discuss this paper on Neuroquery, a relatively new web-based meta-analysis tool: https://elifesciences.org/articles/53385.pdf. This is different from Neurosynth in that it generates meta-analysis maps using predictive modeling from the string of text provided at the prompt, instead of performing inferential statistics to calculate the overlap of activation from different studies. This allows the user to generate predictive maps for more nuanced cognitive processes - especially for clinical populations which may be underrepresented in the literature compared to controls - and can be useful in generating predictions about where the activity will be for one's own study, and for creating ROIs.
Sleep deprivation and the human brain: from brain physiology to cognition”
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.
Interacting spiral wave patterns underlie complex brain dynamics and are related to cognitive processing
The large-scale activity of the human brain exhibits rich and complex patterns, but the spatiotemporal dynamics of these patterns and their functional roles in cognition remain unclear. Here by characterizing moment-by-moment fluctuations of human cortical functional magnetic resonance imaging signals, we show that spiral-like, rotational wave patterns (brain spirals) are widespread during both resting and cognitive task states. These brain spirals propagate across the cortex while rotating around their phase singularity centres, giving rise to spatiotemporal activity dynamics with non-stationary features. The properties of these brain spirals, such as their rotational directions and locations, are task relevant and can be used to classify different cognitive tasks. We also demonstrate that multiple, interacting brain spirals are involved in coordinating the correlated activations and de-activations of distributed functional regions; this mechanism enables flexible reconfiguration of task-driven activity flow between bottom-up and top-down directions during cognitive processing. Our findings suggest that brain spirals organize complex spatiotemporal dynamics of the human brain and have functional correlates to cognitive processing.
Movement planning as a window into hierarchical motor control
The ability to organise one's body for action without having to think about it is taken for granted, whether it is handwriting, typing on a smartphone or computer keyboard, tying a shoelace or playing the piano. When compromised, e.g. in stroke, neurodegenerative and developmental disorders, the individuals’ study, work and day-to-day living are impacted with high societal costs. Until recently, indirect methods such as invasive recordings in animal models, computer simulations, and behavioural markers during sequence execution have been used to study covert motor sequence planning in humans. In this talk, I will demonstrate how multivariate pattern analyses of non-invasive neurophysiological recordings (MEG/EEG), fMRI, and muscular recordings, combined with a new behavioural paradigm, can help us investigate the structure and dynamics of motor sequence control before and after movement execution. Across paradigms, participants learned to retrieve and produce sequences of finger presses from long-term memory. Our findings suggest that sequence planning involves parallel pre-ordering of serial elements of the upcoming sequence, rather than a preparation of a serial trajectory of activation states. Additionally, we observed that the human neocortex automatically reorganizes the order and timing of well-trained movement sequences retrieved from memory into lower and higher-level representations on a trial-by-trial basis. This echoes behavioural transfer across task contexts and flexibility in the final hundreds of milliseconds before movement execution. These findings strongly support a hierarchical and dynamic model of skilled sequence control across the peri-movement phase, which may have implications for clinical interventions.
Mechanisms of human cortical development and neuropsychiatric disease
Internal representation of musical rhythm: transformation from sound to periodic beat
When listening to music, humans readily perceive and move along with a periodic beat. Critically, perception of a periodic beat is commonly elicited by rhythmic stimuli with physical features arranged in a way that is not strictly periodic. Hence, beat perception must capitalize on mechanisms that transform stimulus features into a temporally recurrent format with emphasized beat periodicity. Here, I will present a line of work that aims to clarify the nature and neural basis of this transformation. In these studies, electrophysiological activity was recorded as participants listened to rhythms known to induce perception of a consistent beat across healthy Western adults. The results show that the human brain selectively emphasizes beat representation when it is not acoustically prominent in the stimulus, and this transformation (i) can be captured non-invasively using surface EEG in adult participants, (ii) is already in place in 5- to 6-month-old infants, and (iii) cannot be fully explained by subcortical auditory nonlinearities. Moreover, as revealed by human intracerebral recordings, a prominent beat representation emerges already in the primary auditory cortex. Finally, electrophysiological recordings from the auditory cortex of a rhesus monkey show a significant enhancement of beat periodicities in this area, similar to humans. Taken together, these findings indicate an early, general auditory cortical stage of processing by which rhythmic inputs are rendered more temporally recurrent than they are in reality. Already present in non-human primates and human infants, this "periodized" default format could then be shaped by higher-level associative sensory-motor areas and guide movement in individuals with strongly coupled auditory and motor systems. Together, this highlights the multiplicity of neural processes supporting coordinated musical behaviors widely observed across human cultures.The experiments herein include: a motor timing task comparing the effects of movement vs non-movement with and without feedback (Exp. 1A & 1B), a transcranial magnetic stimulation (TMS) study on the role of the supplementary motor area (SMA) in transforming temporal information (Exp. 2), and a perceptual timing task investigating the effect of noisy movement on time perception with both visual and auditory modalities (Exp. 3A & 3B). Together, the results of these studies support the Bayesian cue combination framework, in that: movement improves the precision of time perception not only in perceptual timing tasks but also motor timing tasks (Exp. 1A & 1B), stimulating the SMA appears to disrupt the transformation of temporal information (Exp. 2), and when movement becomes unreliable or noisy there is no longer an improvement in precision of time perception (Exp. 3A & 3B). Although there is support for the proposed framework, more studies (i.e., fMRI, TMS, EEG, etc.) need to be conducted in order to better understand where and how this may be instantiated in the brain; however, this work provides a starting point to better understanding the intrinsic connection between time and movement
Dynamic endocrine modulation of the nervous system
Sex hormones are powerful neuromodulators of learning and memory. In rodents and nonhuman primates estrogen and progesterone influence the central nervous system across a range of spatiotemporal scales. Yet, their influence on the structural and functional architecture of the human brain is largely unknown. Here, I highlight findings from a series of dense-sampling neuroimaging studies from my laboratory designed to probe the dynamic interplay between the nervous and endocrine systems. Individuals underwent brain imaging and venipuncture every 12-24 hours for 30 consecutive days. These procedures were carried out under freely cycling conditions and again under a pharmacological regimen that chronically suppresses sex hormone production. First, resting state fMRI evidence suggests that transient increases in estrogen drive robust increases in functional connectivity across the brain. Time-lagged methods from dynamical systems analysis further reveals that these transient changes in estrogen enhance within-network integration (i.e. global efficiency) in several large-scale brain networks, particularly Default Mode and Dorsal Attention Networks. Next, using high-resolution hippocampal subfield imaging, we found that intrinsic hormone fluctuations and exogenous hormone manipulations can rapidly and dynamically shape medial temporal lobe morphology. Together, these findings suggest that neuroendocrine factors influence the brain over short and protracted timescales.
More than a beast growing in a passive brain: excitation and inhibition drive epilepsy and glioma progression
Gliomas are brain tumors formed by networks of connected tumor cells, nested in and interacting with neuronal networks. Neuronal activities interfere with tumor growth and occurrence of seizures affects glioma prognosis, while the developing tumor triggers seizures in the infiltrated cortex. Oncometabolites produced by tumor cells and neurotransmitters affect both the generation of epileptic activities by neurons and the growth of glioma cells through synaptic-related mechanisms, involving both GABAergic / Chloride pathways and glutamatergic signaling. From a clinical sight, epilepsy occurrence is beneficial to glioma prognosis but growing tumors are epileptogenic, which constitutes a paradox. This lecture will review how inhibitory and excitatory signaling drives glioma growth and how epileptic and oncological processes are interfering, with a special focus on the human brain.
From cells to systems: multiscale studies of the epileptic brain
It is increasingly recognized that epilepsy affects human brain organization across multiple scales, ranging from cellular alterations in specific regions towards macroscale network imbalances. My talk will overview an emerging paradigm that integrates cellular, neuroimaging, and network modelling approaches to faithful characterize the extent of structural and functional alterations in the common epilepsies. I will also discuss how multiscale framework can help to derive clinically useful biomarkers of dysfunction, and how these methods may guide surgical planning and prognostics.
Retinotopic maps and their relationship to white matter tracts in the human brain
Naturalistic violation of expectations reveal hierarchical surprise responses in the human brain
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.
Cholesterol and matrisome pathways dysregulated in Alzheimer’s disease brain astrocytes and microglia
The impact of apolipoprotein E ε4 (APOE4), the strongest genetic risk factor for Alzheimer’s disease (AD), on human brain cellular function remains unclear. Here, we investigated the effects of APOE4 on brain cell types derived from population and isogenic human induced pluripotent stem cells, post-mortem brain, and APOE targeted replacement mice. Population and isogenic models demonstrate that APOE4 local haplotype, rather than a single risk allele, contributes to risk. Global transcriptomic analyses reveal human-specific, APOE4-driven lipid metabolic dysregulation in astrocytes and microglia. APOE4 enhances de novo cholesterol synthesis despite elevated intracellular cholesterol due to lysosomal cholesterol sequestration in astrocytes. Further, matrisome dysregulation is associated with upregulated chemotaxis, glial activation, and lipid biosynthesis in astrocytes co-cultured with neurons, which recapitulates altered astrocyte matrisome signaling in human brain. Thus, APOE4 initiates glia-specific cell and non-cell autonomous dysregulation that may contribute to increased AD risk." https://doi.org/10.1016/j.cell.2022.05.017
Driving human visual cortex, visually and electrically
The development of circuit-based therapeutics to treat neurological and neuropsychiatric diseases require detailed localization and understanding of electrophysiological signals in the human brain. Electrodes can record and stimulate circuits in many ways, and we often rely on non-invasive imaging methods to predict the location to implant electrodes. However, electrophysiological and imaging signals measure the underlying tissue in a fundamentally different manner. To integrate multimodal data and benefit from these complementary measurements, I will describe an approach that considers how different measurements integrate signals across the underlying tissue. I will show how this approach helps relate fMRI and intracranial EEG measurements and provides new insights into how electrical stimulation influences human brain networks.
Biological and experience-based trajectories in adolescent brain and cognitive development
Adolescent development is not only shaped by the mere passing of time and accumulating experience, but it also depends on pubertal timing and the cascade of maturational processes orchestrated by gonadal hormones. Although individual variability in puberty onset confounds adolescent studies, it has not been efficiently controlled for. Here we introduce ultrasonic bone age assessment to estimate biological maturity and disentangle the independent effects of chronological and biological age on adolescent cognitive abilities, emotional development, and brain maturation. Comparing cognitive performance of participants with different skeletal maturity we uncover the impact of biological age on both IQ and specific abilities. With respect to emotional development, we find narrow windows of highest vulnerability determined by biological age. In terms of neural development, we focus on the relevance of neural states unrelated to sensory stimulation, such as cortical activity during sleep and resting states, and we uncover a novel anterior-to-posterior pattern of human brain maturation. Based on our findings, bone age is a promising biomarker of adolescent maturity.
Trial by trial predictions of subjective time from human brain activity
Our perception of time isn’t like a clock; it varies depending on other aspects of experience, such as what we see and hear in that moment. However, in everyday life, the properties of these simple features can change frequently, presenting a challenge to understanding real-world time perception based on simple lab experiments. We developed a computational model of human time perception based on tracking changes in neural activity across brain regions involved in sensory processing, using fMRI. By measuring changes in brain activity patterns across these regions, our approach accommodates the different and changing feature combinations present in natural scenarios, such as walking on a busy street. Our model reproduces people’s duration reports for natural videos (up to almost half a minute long) and, most importantly, predicts whether a person reports a scene as relatively shorter or longer–the biases in time perception that reflect how natural experience of time deviates from clock time
Zero to Birth: How the Human Brain is Built
By the time a baby is born, its brain is equipped with tens of billions of intricately crafted neurons wired together to form a compact and breathtakingly efficient supercomputer. The book is meant to give a broad audience (i.e. non-neuroscientists) a sense of the step-by-step construction of a human brain as well as our current conceptual understanding of various processes involved. The book also hopes to highlight relevance of brain development to our growing understanding of cognitive and psychological variations and syndromes. The author will talk about the book including the many challenges and rewards involved in writing it.
Hierarchical transformation of visual event timing representations in the human brain: response dynamics in early visual cortex and timing-tuned responses in association cortices
Quantifying the timing (duration and frequency) of brief visual events is vital to human perception, multisensory integration and action planning. For example, this allows us to follow and interact with the precise timing of speech and sports. Here we investigate how visual event timing is represented and transformed across the brain’s hierarchy: from sensory processing areas, through multisensory integration areas, to frontal action planning areas. We hypothesized that the dynamics of neural responses to sensory events in sensory processing areas allows derivation of event timing representations. This would allow higher-level processes such as multisensory integration and action planning to use sensory timing information, without the need for specialized central pacemakers or processes. Using 7T fMRI and neural model-based analyses, we found responses that monotonically increase in amplitude with visual event duration and frequency, becoming increasingly clear from primary visual cortex to lateral occipital visual field maps. Beginning in area MT/V5, we found a gradual transition from monotonic to tuned responses, with response amplitudes peaking at different event timings in different recording sites. While monotonic response components were limited to the retinotopic location of the visual stimulus, timing-tuned response components were independent of the recording sites' preferred visual field positions. These tuned responses formed a network of topographically organized timing maps in superior parietal, postcentral and frontal areas. From anterior to posterior timing maps, multiple events were increasingly integrated, response selectivity narrowed, and responses focused increasingly on the middle of the presented timing range. These results suggest that responses to event timing are transformed from the human brain’s sensory areas to the association cortices, with the event’s temporal properties being increasingly abstracted from the response dynamics and locations of early sensory processing. The resulting abstracted representation of event timing is then propagated through areas implicated in multisensory integration and action planning.
A parsimonious description of global functional brain organization in three spatiotemporal patterns
Resting-state functional magnetic resonance imaging (MRI) has yielded seemingly disparate insights into large-scale organization of the human brain. The brain’s large-scale organization can be divided into two broad categories: zero-lag representations of functional connectivity structure and time-lag representations of traveling wave or propagation structure. In this study, we sought to unify observed phenomena across these two categories in the form of three low-frequency spatiotemporal patterns composed of a mixture of standing and traveling wave dynamics. We showed that a range of empirical phenomena, including functional connectivity gradients, the task-positive/task-negative anti-correlation pattern, the global signal, time-lag propagation patterns, the quasiperiodic pattern and the functional connectome network structure, are manifestations of these three spatiotemporal patterns. These patterns account for much of the global spatial structure that underlies functional connectivity analyses and unifies phenomena in resting-state functional MRI previously thought distinct.
Untitled Seminar
Heiko Luhmann (Germany) – How neuronal activity builds the cerebral cortex; Mary Tolcos (Australia) – Cortical development and fetal brain injury; Silvia Velasco (Australia) – Human brain organoids to study neurodevelopment and disease
A mind set in stone: fossil traces of human brain evolution
Brains do not fossilise, but as they grow and expand during fetal and infant development, they leave an imprint in the bony braincase. Such imprints of fossilised braincases provide direct evidence of brain evolution, but the underlying biological changes have remained elusive. Combining data from fossil skulls, ancient genomes, brain imaging and gene expression helps shed light on the evolutionary changes shaping the human brain. I will highlight two examples separated by more than 3 million years: the evolution of brain growth in Lucy and her kind, and differences between modern humans and Neanderthals.
Chemistry of the adaptive mind: lessons from dopamine
The human brain faces a variety of computational dilemmas, including the flexibility/stability, the speed/accuracy and the labor/leisure tradeoff. I will argue that striatal dopamine is particularly well suited to dynamically regulate these computational tradeoffs depending on constantly changing task demands. This working hypothesis is grounded in evidence from recent studies on learning, motivation and cognitive control in human volunteers, using chemical PET, psychopharmacology, and/or fMRI. These studies also begin to elucidate the mechanisms underlying the huge variability in catecholaminergic drug effects across different individuals and across different task contexts. For example, I will demonstrate how effects of the most commonly used psychostimulant methylphenidate on learning, Pavlovian and effortful instrumental control depend on fluctuations in current environmental volatility, on individual differences in working memory capacity and on opportunity cost respectively.
Can I be bothered? Neural and computational mechanisms underlying the dynamics of effort processing (BACN Early-career Prize Lecture 2021)
From a workout at the gym to helping a colleague with their work, everyday we make decisions about whether we are willing to exert effort to obtain some sort of benefit. Increases in how effortful actions and cognitive processes are perceived to be has been linked to clinically severe impairments to motivation, such as apathy and fatigue, across many neurological and psychiatric conditions. However, the vast majority of neuroscience research has focused on understanding the benefits for acting, the rewards, and not on the effort required. As a result, the computational and neural mechanisms underlying how effort is processed are poorly understood. How do we compute how effortful we perceive a task to be? How does this feed into our motivation and decisions of whether to act? How are such computations implemented in the brain? and how do they change in different environments? I will present a series of studies examining these questions using novel behavioural tasks, computational modelling, fMRI, pharmacological manipulations, and testing in a range of different populations. These studies highlight how the brain represents the costs of exerting effort, and the dynamic processes underlying how our sensitivity to effort changes as a function of our goals, traits, and socio-cognitive processes. This work provides new computational frameworks for understanding and examining impaired motivation across psychiatric and neurological conditions, as well as why all of us, sometimes, can’t be bothered.
Exploring mechanisms of human brain expansion in cerebral organoids
The human brain sets us apart as a species, with its size being one of its most striking features. Brain size is largely determined during development as vast numbers of neurons and supportive glia are generated. In an effort to better understand the events that determine the human brain’s cellular makeup, and its size, we use a human model system in a dish, called cerebral organoids. These 3D tissues are generated from pluripotent stem cells through neural differentiation and a supportive 3D microenvironment to generate organoids with the same tissue architecture as the early human fetal brain. Such organoids are allowing us to tackle questions previously impossible with more traditional approaches. Indeed, our recent findings provide insight into regulation of brain size and neuron number across ape species, identifying key stages of early neural stem cell expansion that set up a larger starting cell number to enable the production of increased numbers of neurons. We are also investigating the role of extrinsic regulators in determining numbers and types of neurons produced in the human cerebral cortex. Overall, our findings are pointing to key, human-specific aspects of brain development and function, that have important implications for neurological disease.
Visualising time in the human brain
We all have a sense of time. Yet it is a particularly intangible sensation. So how is our “sense” of time represented in the brain? Functional neuroimaging studies have consistently identified a network of regions, including Supplementary Motor Area and basal ganglia, that are activated when participants make judgements about the duration of currently unfolding events. In parallel, left parietal cortex and cerebellum are activated when participants predict when future events are likely to occur. These structures are activated by temporal processing even when task goals are purely perceptual. So why should the perception of time be represented in regions of the brain that have more traditionally been implicated in motor function? One possibility is that we learn about time through action. In other words, action could provide the functional scaffolding for learning about time in childhood, explaining why it has come to be represented in motor circuits of the adult brain.
Brain and Mind: Who is the Puppet and who the Puppeteer?
If the mind controls the brain, then there is free will and its corollaries, dignity and responsibility. You are king in your skull-sized kingdom and the architect of your destiny. If, on the other hand, the brain controls the mind, an incendiary conclusion follows: There can be no free will, no praise, no punishment and no purgatory. In this webinar, Professor George Paxinos will discuss his highly respected work on the construction of human and experimental animal brain atlases. He has discovered 94 brain regions, 64 homologies and published 58 books. His first book, The Rat Brain in Stereotaxic Coordinates, is the most cited publication in neuroscience and, for three decades, the third most cited book in science. Professor Paxinos will also present his recently published novel, A River Divided, which was 21 years in the making. Neuroscience principles were used in the formation of charters, such as those related to the mind, soul, free will and consciousness. Environmental issues are at the heart of the novel, including the question of whether the brain is the right ‘size’ for survival. Professor Paxinos studied at Berkeley, McGill and Yale and is now Scientia Professor of Medical Sciences at Neuroscience Research Australia and The University of New South Wales in Sydney.
Rapid learning (and unlearning) in the human brain
Crystallinity characterization of white matter in the human brain
White matter microstructure underpins cognition and function in the human brain through the facilitation of neuronal communication, and the non-invasive characterization of this structure remains an elusive goal in the neuroscience community. Efforts to assess white matter microstructure are hampered by the sheer amount of information needed for characterization. Current techniques address this problem by representing white matter features with single scalars that are often not easy to interpret. Here, we address these issues by introducing tools from soft matter for the characterization of white matter microstructure. We investigate structure on a mesoscopic scale by analyzing its homogeneity and determining which regions of the brain are structurally homogeneous, or ``crystalline" in the context of materials science. We find that crystallinity is a reliable metric that varies across the brain along interpretable lines of anatomical difference. We also parcellate white matter into ``crystal grains," or contiguous sets of voxels of high structural similarity, and find overlap with other white matter parcellations. Our results provide new means of assessing white matter microstructure on multiple length scales, and open new avenues of future inquiry.
Reversing autism-related phenotypes in human brain organoids
The Synaptome Architecture of the Brain: Lifespan, disease, evolution and behavior
The overall aim of my research is to understand how the organisation of the synapse, with particular reference to the postsynaptic proteome (PSP) of excitatory synapses in the brain, informs the fundamental mechanisms of learning, memory and behaviour and how these mechanisms go awry in neurological dysfunction. The PSP indeed bears a remarkable burden of disease, with components being disrupted in disorders (synaptopathies) including schizophrenia, depression, autism and intellectual disability. Our work has been fundamental in revealing and then characterising the unprecedented complexity (>1000 highly conserved proteins) of the PSP in terms of the subsynaptic architecture of postsynaptic proteins such as PSD95 and how these proteins assemble into complexes and supercomplexes in different neurons and regions of the brain. Characterising the PSPs in multiple species, including human and mouse, has revealed differences in key sets of functionally important proteins, correlates with brain imaging and connectome data, and a differential distribution of disease-relevant proteins and pathways. Such studies have also provided important insight into synapse evolution, establishing that vertebrate behavioural complexity is a product of the evolutionary expansion in synapse proteomes that occurred ~500 million years ago. My lab has identified many mutations causing cognitive impairments in mice before they were found to cause human disorders. Our proteomic studies revealed that >130 brain diseases are caused by mutations affecting postsynaptic proteins. We uncovered mechanisms that explain the polygenic basis and age of onset of schizophrenia, with postsynaptic proteins, including PSD95 supercomplexes, carrying much of the polygenic burden. We discovered the “Genetic Lifespan Calendar”, a genomic programme controlling when genes are regulated. We showed that this could explain how schizophrenia susceptibility genes are timed to exert their effects in young adults. The Genes to Cognition programme is the largest genetic study so far undertaken into the synaptic molecular mechanisms underlying behaviour and physiology. We made important conceptual advances that inform how the repertoire of both innate and learned behaviours is built from unique combinations of postsynaptic proteins that either amplify or attenuate the behavioural response. This constitutes a key advance in understanding how the brain decodes information inherent in patterns of nerve impulses, and provides insight into why the PSP has evolved to be so complex, and consequently why the phenotypes of synaptopathies are so diverse. Our most recent work has opened a new phase, and scale, in understanding synapses with the first synaptome maps of the brain. We have developed next-generation methods (SYNMAP) that enable single-synapse resolution molecular mapping across the whole mouse brain and extensive regions of the human brain, revealing the molecular and morphological features of a billion synapses. This has already uncovered unprecedented spatiotemporal synapse diversity organised into an architecture that correlates with the structural and functional connectomes, and shown how mutations that cause cognitive disorders reorganise these synaptome maps; for example, by detecting vulnerable synapse subtypes and synapse loss in Alzheimer’s disease. This innovative synaptome mapping technology has huge potential to help characterise how the brain changes during normal development, including in specific cell types, and with degeneration, facilitating novel pathways to diagnosis and therapy.
Language Representations in the Human Brain: A naturalistic approach
Natural language is strongly context-dependent and can be perceived through different sensory modalities. For example, humans can easily comprehend the meaning of complex narratives presented through auditory speech, written text, or visual images. To understand how complex language-related information is represented in the human brain there is a necessity to map the different linguistic and non-linguistic information perceived under different modalities across the cerebral cortex. To map this information to the brain, I suggest following a naturalistic approach and observing the human brain performing tasks in its naturalistic setting, designing quantitative models that transform real-world stimuli into specific hypothesis-related features, and building predictive models that can relate these features to brain responses. In my talk, I will present models of brain responses collected using functional magnetic resonance imaging while human participants listened to or read natural narrative stories. Using natural text and vector representations derived from natural language processing tools I will present how we can study language processing in the human brain across modalities, in different levels of temporal granularity, and across different languages.
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.
Mapping the Dynamics of the Linear and 3D Genome of Single Cells in the Developing Brain
Three intimately related dimensions of the mammalian genome—linear DNA sequence, gene transcription, and 3D genome architecture—are crucial for the development of nervous systems. Changes in the linear genome (e.g., de novo mutations), transcriptome, and 3D genome structure lead to debilitating neurodevelopmental disorders, such as autism and schizophrenia. However, current technologies and data are severely limited: (1) 3D genome structures of single brain cells have not been solved; (2) little is known about the dynamics of single-cell transcriptome and 3D genome after birth; (3) true de novo mutations are extremely difficult to distinguish from false positives (DNA damage and/or amplification errors). Here, I filled in this longstanding technological and knowledge gap. I recently developed a high-resolution method—diploid chromatin conformation capture (Dip-C)—which resolved the first 3D structure of the human genome, tackling a longstanding problem dating back to the 1880s. Using Dip-C, I obtained the first 3D genome structure of a single brain cell, and created the first transcriptome and 3D genome atlas of the mouse brain during postnatal development. I found that in adults, 3D genome “structure types” delineate all major cell types, with high correlation between chromatin A/B compartments and gene expression. During development, both transcriptome and 3D genome are extensively transformed in the first month of life. In neurons, 3D genome is rewired across scales, correlated with gene expression modules, and independent of sensory experience. Finally, I examined allele-specific structure of imprinted genes, revealing local and chromosome-wide differences. More recently, I expanded my 3D genome atlas to the human and mouse cerebellum—the most consistently affected brain region in autism. I uncovered unique 3D genome rewiring throughout life, providing a structural basis for the cerebellum’s unique mode of development and aging. In addition, to accurately measure de novo mutations in a single cell, I developed a new method—multiplex end-tagging amplification of complementary strands (META-CS), which eliminates nearly all false positives by virtue of DNA complementarity. Using META-CS, I determined the true mutation spectrum of single human brain cells, free from chemical artifacts. Together, my findings uncovered an unknown dimension of neurodevelopment, and open up opportunities for new treatments for autism and other developmental disorders.
Identity-Expression Ambiguity in 3D Morphable Face Models
3D Morphable Models are my favorite class of generative models and are commonly used to model faces. They are typically applied to ill-posed problems such as 3D reconstruction from 2D data. I'll start my presentation with an introduction into 3D Morphable Models and show what they are capable of doing. I'll then focus on our recent finding, the Identity-Expression Ambiguity: We demonstrate that non-orthogonality of the variation in identity and expression can cause identity-expression ambiguity in 3D Morphable Models, and that in practice expression and identity are far from orthogonal and can explain each other surprisingly well. Whilst previously reported ambiguities only arise in an inverse rendering setting, identity-expression ambiguity emerges in the 3D shape generation process itself. The goal of this presentation is to demonstrate the ambiguity and discuss its potential consequences in a computer vision setting as well as for understanding face perception mechanisms in the human brain.