Cortical Circuit
cortical circuit
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
Spike train structure of cortical transcriptomic populations in vivo
The cortex comprises many neuronal types, which can be distinguished by their transcriptomes: the sets of genes they express. Little is known about the in vivo activity of these cell types, particularly as regards the structure of their spike trains, which might provide clues to cortical circuit function. To address this question, we used Neuropixels electrodes to record layer 5 excitatory populations in mouse V1, then transcriptomically identified the recorded cell types. To do so, we performed a subsequent recording of the same cells using 2-photon (2p) calcium imaging, identifying neurons between the two recording modalities by fingerprinting their responses to a “zebra noise” stimulus and estimating the path of the electrode through the 2p stack with a probabilistic method. We then cut brain slices and performed in situ transcriptomics to localize ~300 genes using coppaFISH3d, a new open source method, and aligned the transcriptomic data to the 2p stack. Analysis of the data is ongoing, and suggests substantial differences in spike time coordination between ET and IT neurons, as well as between transcriptomic subtypes of both these excitatory types.
Regulation of cortical circuit maturation and plasticity by oligodendrocytes and myelin
Cell-type-specific plasticity shapes neocortical dynamics for motor learning
How do cortical circuits acquire new dynamics that drive learned movements? This webinar will focus on mouse premotor cortex in relation to learned lick-timing and explore high-density electrophysiology using our silicon neural probes alongside region and cell-type-specific acute genetic manipulations of proteins required for synaptic plasticity.
Blood-brain barrier dysfunction in epilepsy: Time for translation
The neurovascular unit (NVU) consists of cerebral blood vessels, neurons, astrocytes, microglia, and pericytes. It plays a vital role in regulating blood flow and ensuring the proper functioning of neural circuits. Among other, this is made possible by the blood-brain barrier (BBB), which acts as both a physical and functional barrier. Previous studies have shown that dysfunction of the BBB is common in most neurological disorders and is associated with neural dysfunction. Our studies have demonstrated that BBB dysfunction results in the transformation of astrocytes through transforming growth factor beta (TGFβ) signaling. This leads to activation of the innate neuroinflammatory system, changes in the extracellular matrix, and pathological plasticity. These changes ultimately result in dysfunction of the cortical circuit, lower seizure threshold, and spontaneous seizures. Blocking TGFβ signaling and its associated pro-inflammatory pathway can prevent this cascade of events, reduces neuroinflammation, repairs BBB dysfunction, and prevents post-injury epilepsy, as shown in experimental rodents. To further understand and assess BBB integrity in human epilepsy, we developed a novel imaging technique that quantitatively measures BBB permeability. Our findings have confirmed that BBB dysfunction is common in patients with drug-resistant epilepsy and can assist in identifying the ictal-onset zone prior to surgery. Current clinical studies are ongoing to explore the potential of targeting BBB dysfunction as a novel treatment approach and investigate its role in drug resistance, the spread of seizures, and comorbidities associated with epilepsy.
Neuromodulation of striatal D1 cells shapes BOLD fluctuations in anatomically connected thalamic and cortical regions
Understanding how macroscale brain dynamics are shaped by microscale mechanisms is crucial in neuroscience. We investigate this relationship in animal models by directly manipulating cellular properties and measuring whole-brain responses using resting-state fMRI. Specifically, we explore the impact of chemogenetically neuromodulating D1 medium spiny neurons in the dorsomedial caudate putamen (CPdm) on BOLD dynamics within a striato-thalamo-cortical circuit in mice. Our findings indicate that CPdm neuromodulation alters BOLD dynamics in thalamic subregions projecting to the dorsomedial striatum, influencing both local and inter-regional connectivity in cortical areas. This study contributes to understanding structure–function relationships in shaping inter-regional communication between subcortical and cortical levels.
Bio-realistic multiscale modeling of cortical circuits
A central question in neuroscience is how the structure of brain circuits determines their activity and function. To explore this systematically, we developed a 230,000-neuron model of mouse primary visual cortex (area V1). The model integrates a broad array of experimental data:Distribution and morpho-electric properties of different neuron types in V1.
Dynamics of cortical circuits: underlying mechanisms and computational implications
A signature feature of cortical circuits is the irregularity of neuronal firing, which manifests itself in the high temporal variability of spiking and the broad distribution of rates. Theoretical works have shown that this feature emerges dynamically in network models if coupling between cells is strong, i.e. if the mean number of synapses per neuron K is large and synaptic efficacy is of order 1/\sqrt{K}. However, the degree to which these models capture the mechanisms underlying neuronal firing in cortical circuits is not fully understood. Results have been derived using neuron models with current-based synapses, i.e. neglecting the dependence of synaptic current on the membrane potential, and an understanding of how irregular firing emerges in models with conductance-based synapses is still lacking. Moreover, at odds with the nonlinear responses to multiple stimuli observed in cortex, network models with strongly coupled cells respond linearly to inputs. In this talk, I will discuss the emergence of irregular firing and nonlinear response in networks of leaky integrate-and-fire neurons. First, I will show that, when synapses are conductance-based, irregular firing emerges if synaptic efficacy is of order 1/\log(K) and, unlike in current-based models, persists even under the large heterogeneity of connections which has been reported experimentally. I will then describe an analysis of neural responses as a function of coupling strength and show that, while a linear input-output relation is ubiquitous at strong coupling, nonlinear responses are prominent at moderate coupling. I will conclude by discussing experimental evidence of moderate coupling and loose balance in the mouse cortex.
Bridging the gap between artificial models and cortical circuits
Artificial neural networks simplify complex biological circuits into tractable models for computational exploration and experimentation. However, the simplification of artificial models also undermines their applicability to real brain dynamics. Typical efforts to address this mismatch add complexity to increasingly unwieldy models. Here, we take a different approach; by reducing the complexity of a biological cortical culture, we aim to distil the essential factors of neuronal dynamics and plasticity. We leverage recent advances in growing neurons from human induced pluripotent stem cells (hiPSCs) to analyse ex vivo cortical cultures with only two distinct excitatory and inhibitory neuron populations. Over 6 weeks of development, we record from thousands of neurons using high-density microelectrode arrays (HD-MEAs) that allow access to individual neurons and the broader population dynamics. We compare these dynamics to two-population artificial networks of single-compartment neurons with random sparse connections and show that they produce similar dynamics. Specifically, our model captures the firing and bursting statistics of the cultures. Moreover, tightly integrating models and cultures allows us to evaluate the impact of changing architectures over weeks of development, with and without external stimuli. Broadly, the use of simplified cortical cultures enables us to use the repertoire of theoretical neuroscience techniques established over the past decades on artificial network models. Our approach of deriving neural networks from human cells also allows us, for the first time, to directly compare neural dynamics of disease and control. We found that cultures e.g. from epilepsy patients tended to have increasingly more avalanches of synchronous activity over weeks of development, in contrast to the control cultures. Next, we will test possible interventions, in silico and in vitro, in a drive for personalised approaches to medical care. This work starts bridging an important theoretical-experimental neuroscience gap for advancing our understanding of mammalian neuron dynamics.
Investigating activity-dependent processes in cerebral cortex development and disease
The cerebral cortex contains an extraordinary diversity of excitatory projection neuron (PN) and inhibitory interneurons (IN), wired together to form complex circuits. Spatiotemporally coordinated execution of intrinsic molecular programs by PNs and INs and activity-dependent processes, contribute to cortical development and cortical microcircuits formation. Alterations of these delicate processes have often been associated to neurological/neurodevelopmental disorders. However, despite the groundbreaking discovery that spontaneous activity in the embryonic brain can shape regional identities of distinct cortical territories, it is still unclear whether this early activity contributes to define subtype-specific neuronal fate as well as circuit assembly. In this study, we combined in utero genetic perturbations via CRISPR/Cas9 system and pharmacological inhibition of selected ion channels with RNA-sequencing and live imaging technologies to identify the activity-regulated processes controlling the development of different cortical PN classes, their wiring and the acquisition of subtype specific features. Moreover, we generated human induced pluripotent stem cells (iPSCs) form patients affected by a severe, rare and untreatable form of developmental epileptic encephalopathy. By differentiating cortical organoids form patient-derived iPSCs we create human models of early electrical alterations for studying molecular, structural and functional consequences of the genetic mutations during cortical development. Our ultimate goal is to define the activity-conditioned processes that physiologically occur during the development of cortical circuits, to identify novel therapeutical paths to address the pathological consequences of neonatal epilepsies.
From Computation to Large-scale Neural Circuitry in Human Belief Updating
Many decisions under uncertainty entail dynamic belief updating: multiple pieces of evidence informing about the state of the environment are accumulated across time to infer the environmental state, and choose a corresponding action. Traditionally, this process has been conceptualized as a linear and perfect (i.e., without loss) integration of sensory information along purely feedforward sensory-motor pathways. Yet, natural environments can undergo hidden changes in their state, which requires a non-linear accumulation of decision evidence that strikes a tradeoff between stability and flexibility in response to change. How this adaptive computation is implemented in the brain has remained unknown. In this talk, I will present an approach that my laboratory has developed to identify evidence accumulation signatures in human behavior and neural population activity (measured with magnetoencephalography, MEG), across a large number of cortical areas. Applying this approach to data recorded during visual evidence accumulation tasks with change-points, we find that behavior and neural activity in frontal and parietal regions involved in motor planning exhibit hallmarks signatures of adaptive evidence accumulation. The same signatures of adaptive behavior and neural activity emerge naturally from simulations of a biophysically detailed model of a recurrent cortical microcircuit. The MEG data further show that decision dynamics in parietal and frontal cortex are mirrored by a selective modulation of the state of early visual cortex. This state modulation is (i) specifically expressed in the alpha frequency-band, (ii) consistent with feedback of evolving belief states from frontal cortex, (iii) dependent on the environmental volatility, and (iv) amplified by pupil-linked arousal responses during evidence accumulation. Together, our findings link normative decision computations to recurrent cortical circuit dynamics and highlight the adaptive nature of decision-related long-range feedback processing in the brain.
The cortical circuits for hierarchical computation
Heterogeneity and non-random connectivity in reservoir computing
Reservoir computing is a promising framework to study cortical computation, as it is based on continuous, online processing and the requirements and operating principles are compatible with cortical circuit dynamics. However, the framework has issues that limit its scope as a generic model for cortical processing. The most obvious of these is that, in traditional models, learning is restricted to the output projections and takes place in a fully supervised manner. If such an output layer is interpreted at face value as downstream computation, this is biologically questionable. If it is interpreted merely as a demonstration that the network can accurately represent the information, this immediately raises the question of what would be biologically plausible mechanisms for transmitting the information represented by a reservoir and incorporating it in downstream computations. Another major issue is that we have as yet only modest insight into how the structural and dynamical features of a network influence its computational capacity, which is necessary not only for gaining an understanding of those features in biological brains, but also for exploiting reservoir computing as a neuromorphic application. In this talk, I will first demonstrate a method for quantifying the representational capacity of reservoirs without training them on tasks. Based on this technique, which allows systematic comparison of systems, I then present our recent work towards understanding the roles of heterogeneity and connectivity patterns in enhancing both the computational properties of a network and its ability to reliably transmit to downstream networks. Finally, I will give a brief taster of our current efforts to apply the reservoir computing framework to magnetic systems as an approach to neuromorphic computing.
Untitled Seminar
Emilia Favuzzi (USA): Artisans of Brain Wiring: GABA-Receptive Microglia Selectively Sculpt Inhibitory Circuits; Ewoud Schmidt (USA): Humanizing the mouse brain: reorganizing cortical circuits through modified synaptic development; Tracy Bale (USA): Trophoblast mechanisms key in regulating neurodevelopment Anastassia Voronova (Canada): Regulation of neural stem cell fates by neuronal ligands
Restructuring cortical circuits
Neuromodulation of inference and control in the cortical circuits
A biological model system for studying predictive processing
Despite the increasing recognition of predictive processing in circuit neuroscience, little is known about how it may be implemented in cortical circuits. We set out to develop and characterise a biological model system with layer 5 pyramidal cells in the centre. We aim to gain access to prediction and internal model generating processes by controlling, understanding or monitoring everything else: the sensory environment, feed-forward and feed-back inputs, integrative properties, their spiking activity and output. I’ll show recent work from the lab establishing such a model system both in terms of biology as well as tool development.
Taming chaos in neural circuits
Neural circuits exhibit complex activity patterns, both spontaneously and in response to external stimuli. Information encoding and learning in neural circuits depend on the ability of time-varying stimuli to control spontaneous network activity. In particular, variability arising from the sensitivity to initial conditions of recurrent cortical circuits can limit the information conveyed about the sensory input. Spiking and firing rate network models can exhibit such sensitivity to initial conditions that are reflected in their dynamic entropy rate and attractor dimensionality computed from their full Lyapunov spectrum. I will show how chaos in both spiking and rate networks depends on biophysical properties of neurons and the statistics of time-varying stimuli. In spiking networks, increasing the input rate or coupling strength aids in controlling the driven target circuit, which is reflected in both a reduced trial-to-trial variability and a decreased dynamic entropy rate. With sufficiently strong input, a transition towards complete network state control occurs. Surprisingly, this transition does not coincide with the transition from chaos to stability but occurs at even larger values of external input strength. Controllability of spiking activity is facilitated when neurons in the target circuit have a sharp spike onset, thus a high speed by which neurons launch into the action potential. I will also discuss chaos and controllability in firing-rate networks in the balanced state. For these, external control of recurrent dynamics strongly depends on correlations in the input. This phenomenon was studied with a non-stationary dynamic mean-field theory that determines how the activity statistics and the largest Lyapunov exponent depend on frequency and amplitude of the input, recurrent coupling strength, and network size. This shows that uncorrelated inputs facilitate learning in balanced networks. The results highlight the potential of Lyapunov spectrum analysis as a diagnostic for machine learning applications of recurrent networks. They are also relevant in light of recent advances in optogenetics that allow for time-dependent stimulation of a select population of neurons.
Life and death of transient neurons in the development of functional and dysfunctional cortical circuits
Diversification of cortical inhibitory circuits & Molecular programs orchestrating the wiring of inhibitory circuitries
GABAergic interneurons play crucial roles in the regulation of neural activity in the cerebral cortex. In this Dual Lecture, Prof Oscar Marín and Prof Beatriz Rico will discuss several aspects of the formation of inhibitory circuits in the mammalian cerebral cortex. Prof. Marín will provide an overview of the mechanisms regulating the generation of the remarkable diversity of GABAergic interneurons and their ultimate numbers. Prof. Rico will describe the molecular logic through which specific pyramidal cell-interneuron circuits are established in the cerebral cortex, and how alterations in some of these connectivity motifs might be liked to disease. Our web pages for reference: https://devneuro.org.uk/marinlab/ & https://devneuro.org.uk/rico/default
Norepinephrine links astrocytic activity to regulation of cortical state
Cortical state, defined by the synchrony of population-level neuronal activity, is a key determinant of sensory perception. While many arousal-associated neuromodulators—including norepinephrine (NE)—reduce cortical synchrony, how the cortex resynchronizes following NE signaling remains unknown. Using in vivo two-photon imaging and electrophysiology in mouse visual cortex, we describe a critical role for cortical astrocytes in circuit resynchronization. We characterize astrocytes’ sensitive calcium responses to changes in behavioral arousal and NE, identify that astrocyte signaling precedes increases in cortical synchrony, and demonstrate that astrocyte-specific deletion of Adra1A alters arousal-related cortical synchrony. Our findings demonstrate that astrocytic NE signaling acts as a distinct neuromodulatory pathway, regulating cortical state and linking arousal-associated desynchrony to cortical circuit resynchronization.
Hearing in an acoustically varied world
In order for animals to thrive in their complex environments, their sensory systems must form representations of objects that are invariant to changes in some dimensions of their physical cues. For example, we can recognize a friend’s speech in a forest, a small office, and a cathedral, even though the sound reaching our ears will be very different in these three environments. I will discuss our recent experiments into how neurons in auditory cortex can form stable representations of sounds in this acoustically varied world. We began by using a normative computational model of hearing to examine how the brain may recognize a sound source across rooms with different levels of reverberation. The model predicted that reverberations can be removed from the original sound by delaying the inhibitory component of spectrotemporal receptive fields in the presence of stronger reverberation. Our electrophysiological recordings then confirmed that neurons in ferret auditory cortex apply this algorithm to adapt to different room sizes. Our results demonstrate that this neural process is dynamic and adaptive. These studies provide new insights into how we can recognize auditory objects even in highly reverberant environments, and direct further research questions about how reverb adaptation is implemented in the cortical circuit.
A novel form of retinotopy in area V2 highlights location-dependent feature selectivity in the visual system
Topographic maps are a prominent feature of brain organization, reflecting local and large-scale representation of the sensory surface. Traditionally, such representations in early visual areas are conceived as retinotopic maps preserving ego-centric retinal spatial location while ensuring that other features of visual input are uniformly represented for every location in space. I will discuss our recent findings of a striking departure from this simple mapping in the secondary visual area (V2) of the tree shrew that is best described as a sinusoidal transformation of the visual field. This sinusoidal topography is ideal for achieving uniform coverage in an elongated area like V2 as predicted by mathematical models designed for wiring minimization, and provides a novel explanation for stripe-like patterns of intra-cortical connections and functional response properties in V2. Our findings suggest that cortical circuits flexibly implement solutions to sensory surface representation, with dramatic consequences for large-scale cortical organization. Furthermore our work challenges the framework of relatively independent encoding of location and features in the visual system, showing instead location-dependent feature sensitivity produced by specialized processing of different features in different spatial locations. In the second part of the talk, I will propose that location-dependent feature sensitivity is a fundamental organizing principle of the visual system that achieves efficient representation of positional regularities in visual input, and reflects the evolutionary selection of sensory and motor circuits to optimally represent behaviorally relevant information. The relevant papers can be found here: V2 retinotopy (Sedigh-Sarvestani et al. Neuron, 2021) Location-dependent feature sensitivity (Sedigh-Sarvestani et al. Under Review, 2022)
Neurocognitive mechanisms of proactive temporal attention: challenging oscillatory and cortico-centered models
To survive in a rapidly dynamic world, the brain predicts the future state of the world and proactively adjusts perception, attention and action. A key to efficient interaction is to predict and prepare to not only “where” and “what” things will happen, but also to “when”. I will present studies in healthy and neurological populations that investigated the cognitive architecture and neural basis of temporal anticipation. First, influential ‘entrainment’ models suggest that anticipation in rhythmic contexts, e.g. music or biological motion, uniquely relies on alignment of attentional oscillations to external rhythms. Using computational modeling and EEG, I will show that cortical neural patterns previously associated with entrainment in fact overlap with interval timing mechanisms that are used in aperiodic contexts. Second, temporal prediction and attention have commonly been associated with cortical circuits. Studying neurological populations with subcortical degeneration, I will present data that point to a double dissociation between rhythm- and interval-based prediction in the cerebellum and basal ganglia, respectively, and will demonstrate a role for the cerebellum in attentional control of perceptual sensitivity in time. Finally, using EEG in neurodegenerative patients, I will demonstrate that the cerebellum controls temporal adjustment of cortico-striatal neural dynamics, and use computational modeling to identify cerebellar-controlled neural parameters. Altogether, these findings reveal functionally and neural context-specificity and subcortical contributions to temporal anticipation, revising our understanding of dynamic cognition.
The neural basis of pain experience and its modulation by opioids
How the brain creates a painful experience remains a mystery. Solving this mystery is crucial to understanding the fundamental biological processes that underlie the perception of body integrity, and to creating better, non-addictive pain treatments. My laboratory’s goal is to resolve the neural basis of pain. We aim to understand the mechanisms by which our nervous system produces and assembles the sensory-discriminative, affective-motivational, and cognitive-evaluative dimensions of pain to create this unique and critically important experience. To capture every component of the pain experience, we examine the entirety of the pain circuitry, from sensory and spinal ascending pathways to cortical/subcortical circuits and brainstem descending pain modulation systems, at the molecular, cellular, circuit and whole-animal levels. For these studies, we have invented novel behavioral paradigms to interrogate the affective and cognitive dimensions of pain in mice while simultaneously imaging and manipulating nociceptive circuits. My laboratory also investigates how opioids suppress pain. Remarkably, despite their medical and societal significance, how opium poppy alkaloids such as morphine produce profound analgesia remains largely unexplained. By identifying where and how opioids act in neural circuits, we not only establish the mechanisms of action of one of the oldest drugs known to humans, but also reveal the critical elements of the pain circuitry for developing of novel analgesics and bringing an end to the opioid epidemic.
Representation transfer and signal denoising through topographic modularity
To prevail in a dynamic and noisy environment, the brain must create reliable and meaningful representations from sensory inputs that are often ambiguous or corrupt. Since only information that permeates the cortical hierarchy can influence sensory perception and decision-making, it is critical that noisy external stimuli are encoded and propagated through different processing stages with minimal signal degradation. Here we hypothesize that stimulus-specific pathways akin to cortical topographic maps may provide the structural scaffold for such signal routing. We investigate whether the feature-specific pathways within such maps, characterized by the preservation of the relative organization of cells between distinct populations, can guide and route stimulus information throughout the system while retaining representational fidelity. We demonstrate that, in a large modular circuit of spiking neurons comprising multiple sub-networks, topographic projections are not only necessary for accurate propagation of stimulus representations, but can also help the system reduce sensory and intrinsic noise. Moreover, by regulating the effective connectivity and local E/I balance, modular topographic precision enables the system to gradually improve its internal representations and increase signal-to-noise ratio as the input signal passes through the network. Such a denoising function arises beyond a critical transition point in the sharpness of the feed-forward projections, and is characterized by the emergence of inhibition-dominated regimes where population responses along stimulated maps are amplified and others are weakened. Our results indicate that this is a generalizable and robust structural effect, largely independent of the underlying model specificities. Using mean-field approximations, we gain deeper insight into the mechanisms responsible for the qualitative changes in the system’s behavior and show that these depend only on the modular topographic connectivity and stimulus intensity. The general dynamical principle revealed by the theoretical predictions suggest that such a denoising property may be a universal, system-agnostic feature of topographic maps, and may lead to a wide range of behaviorally relevant regimes observed under various experimental conditions: maintaining stable representations of multiple stimuli across cortical circuits; amplifying certain features while suppressing others (winner-take-all circuits); and endow circuits with metastable dynamics (winnerless competition), assumed to be fundamental in a variety of tasks.
Dual lecture: Diversification of cortical inhibitory circuits & Molecular programs orchestrating the wiring of inhibitory circuitries
GABAergic interneurons play crucial roles in the regulation of neural activity in the cerebral cortex. In this Dual Lecture, Prof Oscar Marín and Prof Beatriz Rico will discuss several aspects of the formation of inhibitory circuits in the mammalian cerebral cortex. Prof. Marín will provide an overview of the mechanisms regulating the generation of the remarkable diversity of GABAergic interneurons and their ultimate numbers. Prof. Rico will describe the molecular logic through which specific pyramidal cell-interneuron circuits are established in the cerebral cortex, and how alterations in some of these connectivity motifs might be liked to disease.
Optimising spiking interneuron circuits for compartment-specific feedback
Cortical circuits process information by rich recurrent interactions between excitatory neurons and inhibitory interneurons. One of the prime functions of interneurons is to stabilize the circuit by feedback inhibition, but the level of specificity on which inhibitory feedback operates is not fully resolved. We hypothesized that inhibitory circuits could enable separate feedback control loops for different synaptic input streams, by means of specific feedback inhibition to different neuronal compartments. To investigate this hypothesis, we adopted an optimization approach. Leveraging recent advances in training spiking network models, we optimized the connectivity and short-term plasticity of interneuron circuits for compartment-specific feedback inhibition onto pyramidal neurons. Over the course of the optimization, the interneurons diversified into two classes that resembled parvalbumin (PV) and somatostatin (SST) expressing interneurons. The resulting circuit can be understood as a neural decoder that inverts the nonlinear biophysical computations performed within the pyramidal cells. Our model provides a proof of concept for studying structure-function relations in cortical circuits by a combination of gradient-based optimization and biologically plausible phenomenological models
Deciding to stop deciding: A cortical-subcortical circuit for forming and terminating a decision
The neurobiology of decision-making is informed by neurons capable of representing information over time scales of seconds. Such neurons were initially characterized in studies of spatial working memory, motor planning (e.g., Richard Andersen lab) and spatial attention. For decision-making, such neurons emit graded spike rates, that represent the accumulated evidence for or against a choice. They establish the conduit between the formation of the decision and its completion, usually in the form of a commitment to an action, even if provisional. Indeed, many decisions appear to arise through an accumulation of noisy samples of evidence to a terminating threshold, or bound. Previous studies show that single neurons in the lateral intraparietal area (LIP) represent the accumulation of evidence when monkeys make decisions about the direction of random dot motion (RDM) and express their decision with a saccade to the neuron’s preferred target. The mechanism of termination (the bound) is elusive. LIP is interconnected with other brain regions that also display decision-related activity. Whether these areas play roles in the decision process that are similar to or fundamentally different from that of LIP is unclear. I will present new unpublished experiments that begin to resolve these issues by recording from populations of neurons simultaneously in LIP and one of its primary targets, the superior colliculus (SC), while monkeys make difficult perceptual decisions.
Thalamocortical circuits from neuroanatomy to mental representations
In highly volatile environments, performing actions that address current needs and desires is an ongoing challenge for living organisms. For example, the predictive value of environmental signals needs to be updated when predicted and actual outcomes differ. Furthermore, organisms also need to gain control over the environment through actions that are expected to produce specific outcomes. The data to be presented will show that these processes are highly reliant on thalamocortical circuits wherein thalamic nuclei make a critical contribution to adaptive decision-making, challenging the view that the thalamus only acts as a relay station for the cortical stage. Over the past few years, our work has highlighted the specific contribution of multiple thalamic nuclei in the ability to update the predictive link between events or the causal link between actions and their outcomes via the combination of targeted thalamic interventions (lesion, chemogenetics, disconnections) with behavioral procedures rooted in experimental psychology. We argue that several features of thalamocortical architecture are consistent with a prominent role for thalamic nuclei in shaping mental representations.
Neuronal variability and spatiotemporal dynamics in cortical network models
Neuronal variability is a reflection of recurrent circuitry and cellular physiology. The modulation of neuronal variability is a reliable signature of cognitive and processing state. A pervasive yet puzzling feature of cortical circuits is that despite their complex wiring, population-wide shared spiking variability is low dimensional with all neurons fluctuating en masse. We show that the spatiotemporal dynamics in a spatially structured network produce large population-wide shared variability. When the spatial and temporal scales of inhibitory coupling match known physiology, model spiking neurons naturally generate low dimensional shared variability that captures in vivo population recordings along the visual pathway. Further, we show that firing rate models with spatial coupling can also generate chaotic and low-dimensional rate dynamics. The chaotic parameter region expands when the network is driven by correlated noisy inputs, while being insensitive to the intensity of independent noise.
State-dependent cortical circuits
Spontaneous and sensory-evoked cortical activity is highly state-dependent, promoting the functional flexibility of cortical circuits underlying perception and cognition. Using neural recordings in combination with behavioral state monitoring, we find that arousal and motor activity have complementary roles in regulating local cortical operations, providing dynamic control of sensory encoding. These changes in encoding are linked to altered performance on perceptual tasks. Neuromodulators, such as acetylcholine, may regulate this state-dependent flexibility of cortical network function. We therefore recently developed an approach for dual mesoscopic imaging of acetylcholine release and neural activity across the entire cortical mantle in behaving mice. We find spatiotemporally heterogeneous patterns of cholinergic signaling across the cortex. Transitions between distinct behavioral states reorganize the structure of large-scale cortico-cortical networks and differentially regulate the relationship between cholinergic signals and neural activity. Together, our findings suggest dynamic state-dependent regulation of cortical network operations at the levels of both local and large-scale circuits. Zoom Meeting ID: 964 8138 3003 Contact host if you cannot connect.
Organization and control of hippocampal circuits in epilepsy
Basket cells are key GABAergic inhibitory interneurons that target the somata and proximal dendrites, enabling efficient control of the timing and rate of spiking of their postsynaptic targets. In all cortical circuits, there are two major types of basket cell that exhibit striking developmental, molecular, anatomical, and physiological differences. In this talk, I will discuss recent results that reveal the tightly coupled complementarity of these two key microcircuit regulatory modules, demonstrating a novel form of brain-state-specific segregation of inhibition during spontaneous behavior, with implications for the assessment of dysregulated inhibition in epilepsy. In addition, I will describe recent advances in our understanding of the spatio-temporal dynamics of endocannabinoid signaling in hippocampal circuits and discuss how abnormal amplification of these activity-dependent signaling processes leads to surprising downstream effects in seizures.
Cortical and sub-cortical circuits for auditory perception and learning
Sonic hedgehog signaling: from neurons to astrocytes during cortical circuit assembly
A Cortical Circuit for Audio-Visual Predictions
Team work makes sensory streams work: our senses work together, learn from each other, and stand in for one another, the result of which is perception and understanding. Learned associations between stimuli in different sensory modalities can shape the way we perceive these stimuli (Mcgurk and Macdonald, 1976). During audio-visual associative learning, auditory cortex is thought to underlie multi-modal plasticity in visual cortex (McIntosh et al., 1998; Mishra et al., 2007; Zangenehpour and Zatorre, 2010). However, it is not well understood how processing in visual cortex is altered by an auditory stimulus that is predictive of a visual stimulus and what the mechanisms are that mediate such experience-dependent, audio-visual associations in sensory cortex. Here we describe a neural mechanism by which an auditory input can shape visual representations of behaviorally relevant stimuli through direct interactions between auditory and visual cortices. We show that the association of an auditory stimulus with a visual stimulus in a behaviorally relevant context leads to an experience-dependent suppression of visual responses in primary visual cortex (V1). Auditory cortex axons carry a mixture of auditory and retinotopically-matched visual input to V1, and optogenetic stimulation of these axons selectively suppresses V1 neurons responsive to the associated visual stimulus after, but not before, learning. Our results suggest that cross-modal associations can be stored in long-range cortical connections and that with learning these cross-modal connections function to suppress the responses to predictable input.
Transient cortical circuits match spontaneous and sensory driven activity during development
Cortical networks for flexible decisions during spatial navigation
My lab seeks to understand how the mammalian brain performs the computations that underlie cognitive functions, including decision-making, short-term memory, and spatial navigation, at the level of the building blocks of the nervous system, cell types and neural populations organized into circuits. We have developed methods to measure, manipulate, and analyze neural circuits across various spatial and temporal scales, including technology for virtual reality, optical imaging, optogenetics, intracellular electrophysiology, molecular sensors, and computational modeling. I will present recent work that uses large scale calcium imaging to reveal the functional organization of the mouse posterior cortex for flexible decision-making during spatial navigation in virtual reality. I will also discuss work that uses optogenetics and calcium imaging during a variety of decision-making tasks to highlight how cognitive experience and context greatly alter the cortical circuits necessary for navigation decisions.
Playing fast and loose with glutamate builds healthy circuits in the developing cortex
The construction of cortical circuits requires the precise formation of connections between excitatory and inhibitory neurons during early development. Multiple factors, including neurotransmitters, neuronal activity, and neuronal-glial interactions, shape how these critical circuits form. Disruptions of these early processes can disrupt circuit formation, leading to epilepsy and other neurodevelopmental disorders. Here, I will describe our work into understanding how prolonged post-natal astrocyte development in the cortex creates a permissive window for glutamate signaling that provides tonic activation of developing interneurons through Grin2D NMDA receptors. Experimental disruption of this pathway results in hyperexcitable cortical circuits and human mutations in the Grin2D gene, as well as other related molecules that regulate early life glutamate signaling, are associated with devastating epileptic encephalopathies. We will explore fundamental mechanisms linking early life glutamate signaling and later circuit hyperexcitability, with an emphasis on potential therapeutic interventions aimed at reducing epilepsy and other neurological dysfunction.
The emergence and modulation of time in neural circuits and behavior
Spontaneous behavior in animals and humans shows a striking amount of variability both in the spatial domain (which actions to choose) and temporal domain (when to act). Concatenating actions into sequences and behavioral plans reveals the existence of a hierarchy of timescales ranging from hundreds of milliseconds to minutes. How do multiple timescales emerge from neural circuit dynamics? How do circuits modulate temporal responses to flexibly adapt to changing demands? In this talk, we will present recent results from experiments and theory suggesting a new computational mechanism generating the temporal variability underlying naturalistic behavior and cortical activity. We will show how neural activity from premotor areas unfolds through temporal sequences of attractors, which predict the intention to act. These sequences naturally emerge from recurrent cortical networks, where correlated neural variability plays a crucial role in explaining the observed variability in action timing. We will then discuss how reaction times can be accelerated or slowed down via gain modulation, flexibly induced by neuromodulation or perturbations; and how gain modulation may control response timing in the visual cortex. Finally, we will present a new biologically plausible way to generate a reservoir of multiple timescales in cortical circuits.
State-dependent cortical circuits
Spontaneous and sensory-evoked cortical activity is highly state-dependent, promoting the functional flexibility of cortical circuits underlying perception and cognition. Using neural recordings in combination with behavioral state monitoring, we find that arousal and motor activity have complementary roles in regulating local cortical operations, providing dynamic control of sensory encoding. These changes in encoding are linked to altered performance on perceptual tasks. Neuromodulators, such as acetylcholine, may regulate this state-dependent flexibility of cortical network function. We therefore recently developed an approach for dual mesoscopic imaging of acetylcholine release and neural activity across the entire cortical mantle in behaving mice. We find spatiotemporally heterogeneous patterns of cholinergic signaling across the cortex. Transitions between distinct behavioral states reorganize the structure of large-scale cortico-cortical networks and differentially regulate the relationship between cholinergic signals and neural activity. Together, our findings suggest dynamic state-dependent regulation of cortical network operations at the levels of both local and large-scale circuits.
Cortical interneuron wiring in health and disease
The establishment of synaptic connections is essential for normal brain function, yet the molecular mechanisms responsible for the precise connectivity of specific neural circuits remain largely unknown. Previous work has shown that the assembly of cortical circuits requires specific functions of molecular signalling complexes at different classes of synapses. In this talk, I will describe the molecular logic through which specific pyramidal cell-interneuron circuits are established in the cerebral cortex of the mouse, and how alterations in some of these connectivity motifs might be liked to disease.
How does the cortex integrate conflicting time-information? A model of temporal averaging
In daily life, we consistently make decisions in pursuit of some goal. Many decisions are informed by multiple sources of information. Unfortunately, these sources often provide ambiguous information about what course of action to take. Therefore, determining how the brain integrates information to resolve this ambiguity is key to understanding the neural mechanisms of decision-making. In the domain of time, this topic can be studied by training subjects to predict when a future event will occur based on distinct cues (e.g., tone, light, etc.). If multiple cues are presented simultaneously and their cue-to-event intervals differ (e.g., tone-10s + light-30s), subjects will often expect the event to occur at the average of their intervals. This ‘temporal averaging’ effect is presumably how the timing system resolves ambiguous time-information. The neural mechanisms of temporal averaging are currently unclear. Here, we will propose how temporal averaging could emerge in cortical circuits using a simple modification of a ‘drift-diffusion’ model of timing.
In vivo modelling of human cortical circuit wiring and dysfunction
Dimensions of variability in circuit models of cortex
Cortical circuits receive multiple inputs from upstream populations with non-overlapping stimulus tuning preferences. Both the feedforward and recurrent architectures of the receiving cortical layer will reflect this diverse input tuning. We study how population-wide neuronal variability propagates through a hierarchical cortical network receiving multiple, independent, tuned inputs. We present new analysis of in vivo neural data from the primate visual system showing that the number of latent variables (dimension) needed to describe population shared variability is smaller in V4 populations compared to those of its downstream visual area PFC. We successfully reproduce this dimensionality expansion from our V4 to PFC neural data using a multi-layer spiking network with structured, feedforward projections and recurrent assemblies of multiple, tuned neuron populations. We show that tuning-structured connectivity generates attractor dynamics within the recurrent PFC current, where attractor competition is reflected in the high dimensional shared variability across the population. Indeed, restricting the dimensionality analysis to activity from one attractor state recovers the low-dimensional structure inherited from each of our tuned inputs. Our model thus introduces a framework where high-dimensional cortical variability is understood as ``time-sharing’’ between distinct low-dimensional, tuning-specific circuit dynamics.
The emergence of contrast invariance in cortical circuits
Neurons in the primary visual cortex (V1) encode the orientation and contrast of visual stimuli through changes in firing rate (Hubel and Wiesel, 1962). Their activity typically peaks at a preferred orientation and decays to zero at the orientations that are orthogonal to the preferred. This activity pattern is re-scaled by contrast but its shape is preserved, a phenomenon known as contrast invariance. Contrast-invariant selectivity is also observed at the population level in V1 (Carandini and Sengpiel, 2004). The mechanisms supporting the emergence of contrast-invariance at the population level remain unclear. How does the activity of different neurons with diverse orientation selectivity and non-linear contrast sensitivity combine to give rise to contrast-invariant population selectivity? Theoretical studies have shown that in the balance limit, the properties of single-neurons do not determine the population activity (van Vreeswijk and Sompolinsky, 1996). Instead, the synaptic dynamics (Mongillo et al., 2012) as well as the intracortical connectivity (Rosenbaum and Doiron, 2014) shape the population activity in balanced networks. We report that short-term plasticity can change the synaptic strength between neurons as a function of the presynaptic activity, which in turns modifies the population response to a stimulus. Thus, the same circuit can process a stimulus in different ways –linearly, sublinearly, supralinearly – depending on the properties of the synapses. We found that balanced networks with excitatory to excitatory short-term synaptic plasticity cannot be contrast-invariant. Instead, short-term plasticity modifies the network selectivity such that the tuning curves are narrower (broader) for increasing contrast if synapses are facilitating (depressing). Based on these results, we wondered whether balanced networks with plastic synapses (other than short-term) can support the emergence of contrast-invariant selectivity. Mathematically, we found that the only synaptic transformation that supports perfect contrast invariance in balanced networks is a power-law release of neurotransmitter as a function of the presynaptic firing rate (in the excitatory to excitatory and in the excitatory to inhibitory neurons). We validate this finding using spiking network simulations, where we report contrast-invariant tuning curves when synapses release the neurotransmitter following a power- law function of the presynaptic firing rate. In summary, we show that synaptic plasticity controls the type of non-linear network response to stimulus contrast and that it can be a potential mechanism mediating the emergence of contrast invariance in balanced networks with orientation-dependent connectivity. Our results therefore connect the physiology of individual synapses to the network level and may help understand the establishment of contrast-invariant selectivity.
State-dependent regulation of cortical circuits
Spontaneous and sensory-evoked cortical activity is highly state-dependent, promoting the functional flexibility of cortical circuits underlying perception and cognition. Using neural recordings in combination with behavioral state monitoring, we find that arousal and motor activity have complementary roles in regulating local cortical operations, providing dynamic control of sensory encoding. These changes in encoding are linked to altered performance on perceptual tasks. Neuromodulators, such as acetylcholine, may regulate this state-dependent flexibility of cortical network function. We therefore recently developed an approach for dual mesoscopic imaging of acetylcholine release and neural activity across the entire cortical mantle in behaving mice. We find spatiotemporally heterogeneous patterns of cholinergic signaling across the cortex. Transitions between distinct behavioral states reorganize the structure of large-scale cortico-cortical networks and differentially regulate the relationship between cholinergic signals and neural activity. Together, our findings suggest dynamic state-dependent regulation of cortical network operations at the levels of both local and large-scale circuits.
Circuit mechanisms underlying the dynamic control of cortical processing by subcortical neuromodulators
Behavioral states such as arousal and attention can have profound effects on sensory processing, determining how – sometimes whether – a stimulus is processed. This state-dependence is believed to arise, at least in part, as a result of inputs to cortex from subcortical structures that release neuromodulators such as acetylcholine, noradrenaline, and serotonin, often non-synaptically. The mechanisms that underlie the interaction between these “wireless” non-synaptic signals and the “wired” cortical circuit are not well understood. Furthermore, neuromodulatory signaling is traditionally considered broad in its impact across cortex (within a species) and consistent in its form and function across species (at least in mammals). The work I will present approaches the challenge of understanding neuromodulatory action in the cortex from a number of angles: anatomy, physiology, pharmacology, and chemistry. The overarching goal of our effort is to elucidate the mechanisms behind local neuromodulation in the cortex of non-human primates, and to reveal differences in structure and function across cortical model systems.
Thalamocortical circuits from neuroanatomy to cognitive processes
Towards multipurpose biophysics-based mathematical models of cortical circuits
Starting with the work of Hodgkin and Huxley in the 1950s, we now have a fairly good understanding of how the spiking activity of neurons can be modelled mathematically. For cortical circuits the understanding is much more limited. Most network studies have considered stylized models with a single or a handful of neuronal populations consisting of identical neurons with statistically identical connection properties. However, real cortical networks have heterogeneous neural populations and much more structured synaptic connections. Unlike typical simplified cortical network models, real networks are also “multipurpose” in that they perform multiple functions. Historically the lack of computational resources has hampered the mathematical exploration of cortical networks. With the advent of modern supercomputers, however, simulations of networks comprising hundreds of thousands biologically detailed neurons are becoming feasible (Einevoll et al, Neuron, 2019). Further, a large-scale biologically network model of the mouse primary visual cortex comprising 230.000 neurons has recently been developed at the Allen Institute for Brain Science (Billeh et al, Neuron, 2020). Using this model as a starting point, I will discuss how we can move towards multipurpose models that incorporate the true biological complexity of cortical circuits and faithfully reproduce multiple experimental observables such as spiking activity, local field potentials or two-photon calcium imaging signals. Further, I will discuss how such validated comprehensive network models can be used to gain insights into the functioning of cortical circuits.
Autism-Associated Shank3 Is Essential for Homeostatic Compensation in Rodent Visual Cortex
Neocortical networks must generate and maintain stable activity patterns despite perturbations induced by learning and experience- dependent plasticity. There is abundant theoretical and experimental evidence that network stability is achieved through homeostatic plasticity mechanisms that adjust synaptic and neuronal properties to stabilize some measure of average activity, and this process has been extensively studied in primary visual cortex (V1), where chronic visual deprivation induces an initial drop in activity and ensemble average firing rates (FRs), but over time activity is restored to baseline despite continued deprivation. Here I discuss recent work from the lab in which we followed this FR homeostasis in individual V1 neurons in freely behaving animals during a prolonged visual deprivation/eye-reopening paradigm. We find that - when FRs are perturbed by manipulating sensory experience - over time they return precisely to a cell-autonomous set-point. Finally, we find that homeostatic plasticity is perturbed in a mouse model of Autism spectrum disorder, and this results in a breakdown of FRH within V1. These data suggest that loss of homeostatic plasticity is one primary cause of excitation/inhibition imbalances in ASD models. Together these studies illuminate the role of stabilizing plasticity mechanisms in the ability of neocortical circuits to recover robust function following challenges to their excitability.
Predictive processing in cortical circuits
Misplaced and misconnected: circuit-level defects in malformations of cortical development
During histogenesis of the cerebral cortex, a proper laminar placement of defined numbers of specific cellular types is necessary to ensure proper functional connectivity patterns. There is a wide range of cortical malformations causing epilepsy and intellectual disability in humans, characterized with various degrees of neuronal misplacement, aberrant circuit organization or abnormal folding patterns. Although progress in human neurogenetics and brain imaging techniques have considerably advanced the identification of their causative genes, the pathophysiological mechanisms associated with defective cerebral cortex development remain poorly understood. In my presentation, I will outline some of our recent works in rodent models illustrating how misplaced neurons forming grey matter heterotopia, a cortical malformation subtype, interfere with the proper development of cortical circuits, and induce both local and distant circuitry changes associated with the subsequent emergence of epilepsy.
Neural and computational principles of the processing of dynamic faces and bodies
Body motion is a fundamental signal of social communication. This includes facial as well as full-body movements. Combining advanced methods from computer animation with motion capture in humans and monkeys, we synthesized highly-realistic monkey avatar models. Our face avatar is perceived by monkeys as almost equivalent to a real animal, and does not induce an ‘uncanny valley effect’, unlike all other previously used avatar models in studies with monkeys. Applying machine-learning methods for the control of motion style, we were able to investigate how species-specific shape and dynamic cues influence the perception of human and monkey facial expressions. Human observers showed very fast learning of monkey expressions, and a perceptual encoding of expression dynamics that was largely independent of facial shape. This result is in line with the fact that facial shape evolved faster than the neuromuscular control in primate phylogenesis. At the same time, it challenges popular neural network models of the recognition of dynamic faces that assume a joint encoding of facial shape and dynamics. We propose an alternative physiologically-inspired neural model that realizes such an orthogonal encoding of facial shape and expression from video sequences. As second example, we investigated the perception of social interactions from abstract stimuli, similar to the ones by Heider & Simmel (1944), and also from more realistic stimuli. We developed and validated a new generative model for the synthesis of such social interaction, which is based on a modification of human navigation model. We demonstrate that the recognition of such stimuli, including the perception of agency, can be accounted for by a relatively elementary physiologically-inspired hierarchical neural recognition model, that does not require the assumption of sophisticated inference mechanisms, as postulated by some cognitive theories of social recognition. Summarizing, this suggests that essential phenomena in social cognition might be accounted for by a small set of simple neural principles that can be easily implemented by cortical circuits. The developed technologies for stimulus control form the basis of electrophysiological studies that can verify specific neural circuits, as the ones proposed by our theoretical models.
Microglia and neuroimmune interactions in the wiring of cortical circuits
Hippocampal disinhibitory circuits: cell types, connectivity and function
The concept of a dynamic excitation / inhibition ratio, that can shape information flow in cortical circuits during complex behavioural tasks due to circuit disinhibition, has recently arisen as an important and conserved processing motif. It has been also recognized that, in cortical circuits, a subpopulation of GABAergic cells that express vasoactive intestinal polypeptide (VIP) innervates selectively inhibitory interneurons, providing for circuit disinhibition as a possible outcome, depending on the network state and behavioural context. In this talk, I will highlight the latest discoveries on the dynamic organization of hippocampal disinhibitory circuits with a focus on VIP-expressing interneurons. I will discuss the neuron types that can be involved in disinhibition and their local circuit and long-range synaptic connections. I will also discuss some recent findings on how hippocampal VIP circuits may coordinate spatial learning.
Cortical circuits for olfactory navigation
Olfactory navigation is essential for the survival of living beings from unicellular organisms to mammals. In the wild, rodents combine odor information with an internal spatial representation of the environment for foraging and navigation. What are the neural circuits in the brain that implement these behaviours? My research addresses this question by examining the synaptic circuits and neural population activity in the olfactory cortex to understand the integration of olfactory and spatial information. Primary olfactory (piriform) cortex (PCx) has long been recognized as a highly associative brain structure. What is the behavioural and functional role of these associative synapses in PCx? We designed an odor-cued navigation task, where rats must use both olfactory and spatial information to obtain water rewards. We recorded from populations of posterior piriform cortex (pPCx) neurons during behaviour and found that individual neurons were not only odor-selective, but also fired differentially to the same odor sampled at different locations, forming an “olfactory place map”. Spatial locations can be decoded from simultaneously recorded pPCx population, and spatial selectivity is maintained in the absence of odors, across behavioural contexts. This novel olfactory place map is consistent with our finding for a dominant role of associative excitatory synapses in shaping PCx representations, and suggest a role for PCx spatial representations in supporting olfactory navigation. This work not only provides insight into the neural basis for how odors can be used for navigation, but also reveals PCx as a prime site for addressing the general question of how sensory information is anchored within memory systems and combined with cognitive maps to guide flexible behaviour.
Playing the piano with the cortex: role of neuronal ensembles and pattern completion in perception
The design of neural circuits, with large numbers of neurons interconnected in vast networks, strongly suggest that they are specifically build to generate emergent functional properties (1). To explore this hypothesis, we have developed two-photon holographic methods to selective image and manipulate the activity of neuronal populations in 3D in vivo (2). Using them we find that groups of synchronous neurons (neuronal ensembles) dominate the evoked and spontaneous activity of mouse primary visual cortex (3). Ensembles can be optogenetically imprinted for several days and some of their neurons trigger the entire ensemble (4). By activating these pattern completion cells in ensembles involved in visual discrimination paradigms, we can bi-directionally alter behavioural choices (5). Our results demonstrate that ensembles are necessary and sufficient for visual perception and are consistent with the possibility that neuronal ensembles are the functional building blocks of cortical circuits. 1. R. Yuste, From the neuron doctrine to neural networks. Nat Rev Neurosci 16, 487-497 (2015). 2. L. Carrillo-Reid, W. Yang, J. E. Kang Miller, D. S. Peterka, R. Yuste, Imaging and Optically Manipulating Neuronal Ensembles. Annu Rev Biophys, 46: 271-293 (2017). 3. J. E. Miller, I. Ayzenshtat, L. Carrillo-Reid, R. Yuste, Visual stimuli recruit intrinsically generated cortical ensembles. Proceedings of the National Academy of Sciences of the United States of America 111, E4053-4061 (2014). 4. L. Carrillo-Reid, W. Yang, Y. Bando, D. S. Peterka, R. Yuste, Imprinting and recalling cortical ensembles. Science 353, 691-694 (2016). 5. L. Carrillo-Reid, S. Han, W. Yang, A. Akrouh, R. Yuste, (2019). Controlling visually-guided behaviour by holographic recalling of cortical ensembles. Cell 178, 447-457. DOI:https://doi.org/10.1016/j.cell.2019.05.045.
A human-specific modifier of synaptic development, cortical circuit connectivity and function
The remarkable cognitive abilities characterizing humans has been linked to unique patterns of connectivity characterizing the neocortex. Comparative studies have shown that human cortical pyramidal neurons (PN) receive a significant increase of synaptic inputs when compared to other mammals, including non-human primates and rodents, but how this may relate to changes in cortical connectivity and function remained largely unknown. We previously identified a human-specific gene duplication (HSGD), SRGAP2C, that, when induced in mouse cortical PNs drives human-specific features of synaptic development, including a correlated increase in excitatory (E) and inhibitory (I) synapse density through inhibition of the ancestral SRGAP2A protein (Charrier et al. 2012; Fossatti et al. 2016; Schmidt et al. 2019). However, the origin and nature of this increased connectivity and its impact on cortical circuit function was unknown. I will present new results exploring these questions (see Schmidt et al. (2020) https://www.biorxiv.org/content/10.1101/852970v1). Using a combination of transgenic approaches and quantitative monosynaptic tracing, we discovered that humanization of SRGAP2C expression in the mouse cortex leads to a specific increase in local and long-range cortico-cortical inputs received by layer 2/3 cortical PNs. Moreover, using in vivo two-photon imaging in the barrel cortex of awake mice, we show that humanization of SRGAP2C expression increases the reliability and selectivity of sensory- evoked responses in layer 2/3 PNs. We also found that mice humanized for SRGAP2C in all cortical pyramidal neurons and throughout development are characterized by improved behavioural performance in a novel whisker-based sensory discrimination task compared to control wild-type mice. Our results suggest that the emergence of SRGAP2C during human evolution underlie a new substrate for human brain evolution whereby it led to increased local and long-range cortico-cortical connectivity and improved reliability of sensory-evoked cortical coding. References cited Charrier C.*, Joshi K. *, Coutinho-Budd J., Kim, J-E., Lambert N., de Marchena, J., Jin W-L., Vanderhaeghen P., Ghosh A., Sassa T, and Polleux F. (2012) Inhibition of SRGAP2 function by its human-specific paralogs induces neoteny of spine maturation. Cell 149:923-935. * Co-first authors. Fossati M, Pizzarelli R, Schmidt ER, Kupferman JV, Stroebel D, Polleux F*, Charrier C*. (2016) SRGAP2 and Its Human-Specific Paralog Co-Regulate the Development of Excitatory and Inhibitory Synapses. Neuron. 91(2):356-69. * Co-senior corresponding authors. Schmidt E.R.E., Kupferman J.V., Stackmann M., Polleux F. (2019) The human-specific paralogs SRGAP2 and SRGAP2C differentially modulate SRGAP2A-dependent synaptic development. Scientific Rep. 9(1):18692. Schmidt E.R.E, Zhao H.T., Hillman E.M.C., Polleux F. (2020) Humanization of SRGAP2C expression increases cortico-cortical connectivity and reliability of sensory-evoked responses in mouse brain. Submitted. See also: https://www.biorxiv.org/content/10.1101/852970v1
Fate and freedom in developing neocortical circuits
During brain development, neurons are born in specialized niches and migrate to target regions where they assemble to form the circuits that underlie mammalian behaviour. During their journey, neurons follow cell-intrinsic, genetic programs transmitted by their mother cells but also environmental cues, which together drive their maturation. Here, focusing on the neocortex, I will discuss recent findings from our laboratory in which we untangle and manipulate the programs at play in progenitors and their daughter neurons to better understand the emergence of cellular diversity in the developing brain.
Identifying plasticity mechanisms underlying experience-driven adaptation in cortical circuits
Bernstein Conference 2024
Top-down modulation shapes timescales via synaptic plasticity in cortical circuits with multiple interneuron types
Bernstein Conference 2024
Top-down modulation in canonical cortical circuits with inhibitory short-term plasticity
COSYNE 2022
Top-down modulation in canonical cortical circuits with inhibitory short-term plasticity
COSYNE 2022
Connectome-constrained cortical circuits optimized for visual function and working memory tasks
COSYNE 2023
Recurrent circuits improve neural response prediction and provide insight into cortical circuits
COSYNE 2023
State-dependent modulation of dependencies in a laminar cortical circuit
COSYNE 2023
A thalamocortical circuit gates memory consolidation
COSYNE 2023
Unifying mechanistic and functional models of cortical circuits with low-rank, E/I-balanced spiking networks
COSYNE 2023
Inhibitory plasticity-based stabilization of cortical circuits predicts novel paradoxical effects
COSYNE 2025
Inhibitory cell-type-specific properties and transformations set up a unique depth-dependent temporal integration scheme in the human neocortical circuit
COSYNE 2025
Top-down modulation shapes the timescales of cortical circuits via synaptic plasticity
COSYNE 2025
BACE1-dependent effects of IL-6 on cortical circuits
FENS Forum 2024
Carbon-based neural interfaces to probe retinal and cortical circuits with functional ultrasound imaging in vivo
FENS Forum 2024
Control of cortical circuits by layer 1 NDNF interneurons
FENS Forum 2024
Cortical circuits for context dependent sensory processing
FENS Forum 2024
Cortical circuits for goal-directed cross-modal transfer learning
FENS Forum 2024
Dynamics of focally hyperconcentrated drug release for non-invasive cortical circuit manipulation
FENS Forum 2024
Harmonic oscillator networks (HORNs) and the functional role of oscillatory dynamics in neocortical circuits
FENS Forum 2024
Reorganisation of modular activity in cortical circuits
FENS Forum 2024
The role of TCF7L2 transcription factor in the function of thalamocortical circuits
FENS Forum 2024