1
|
Piña Novo D, Gao M, Fischer R, Richevaux L, Yu J, Barrett JM, Shepherd GMG. Cortical dynamics in hand/forelimb S1 and M1 evoked by brief photostimulation of the mouse's hand. eLife 2025; 14:RP105112. [PMID: 40387088 PMCID: PMC12088680 DOI: 10.7554/elife.105112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025] Open
Abstract
Spiking activity along synaptic circuits linking primary somatosensory (S1) and motor (M1) areas is fundamental for sensorimotor integration in cortex. Circuits along the ascending somatosensory pathway through mouse hand/forelimb S1 and M1 were recently described in detail (Yamawaki et al., 2021). Here, we characterize the peripherally evoked spiking dynamics in these two cortical areas. Brief (5 ms) optogenetic photostimulation of the hand generated short (~25 ms) barrages of activity first in S1 (onset latency 15 ms) then M1 (10 ms later). The estimated propagation speed was 20-fold faster from hand to S1 than from S1 to M1. Amplitudes in M1 were strongly attenuated. Responses were typically triphasic, with suppression and rebound following the initial peak. Evoked activity in S1 was biased to middle layers, consistent with thalamocortical connectivity, while that in M1 was biased to upper layers, consistent with corticocortical connectivity. Parvalbumin (PV) inhibitory interneurons were involved in each phase, accounting for three quarters of the initial spikes generated in S1, and their selective photostimulation sufficed to evoke suppression and rebound in both S1 and M1. Partial silencing of S1 by PV activation during hand stimulation reduced the M1 sensory responses. Overall, these results characterize how evoked spiking activity propagates along the hand/forelimb transcortical loop, and illuminate how in vivo cortical dynamics relate to the underlying synaptic circuit organization in this system.
Collapse
Affiliation(s)
- Daniela Piña Novo
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Mang Gao
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Rita Fischer
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Louis Richevaux
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Jianing Yu
- School of Life Sciences, Peking UniversityBeijingChina
| | - John M Barrett
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Gordon MG Shepherd
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| |
Collapse
|
2
|
Paparella I, Cardone P, Zanichelli B, Lamalle L, Collette F, Sherif S, Zubkov M, Clarke WT, Stagg CJ, Maquet P, Vandewalle G. 7 Tesla magnetic resonance spectroscopy estimates of GABA concentration relate to physiological measures of tonic inhibition in the human motor cortex. J Physiol 2025; 603:2821-2838. [PMID: 40173216 DOI: 10.1113/jp287311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 03/18/2025] [Indexed: 04/04/2025] Open
Abstract
GABAergic neurotransmission within the cortex plays a key role in learning and is altered in several brain diseases. Quantification of bulk GABA in the human brain is typically obtained by magnetic resonance spectroscopy (MRS). However, the interpretation of MRS-GABA is still debated. A recent mathematical simulation contends that MRS detects extrasynaptic GABA, mediating tonic inhibition. Nevertheless, no empirical data have yet confirmed this hypothesis. Here we collected ultra-high-field 7 Tesla MRS and transcranial magnetic stimulation coupled with high-density electroencephalography (TMS-hdEEG) from the motor cortex of 20 healthy participants (age 23.95 ± 6.4 years), while they were at rest. We first applied a neural mass model (NMM) to TMS-evoked potentials to disentangle the contribution of different GABAergic pools. We then assessed to which of these different pools MRS-GABA was related to by means of parametric empirical Bayesian (PEB) analysis. We found that MRS-GABA was mostly positively related to the NMM-derived measures of tonic inhibition and overall functionality of the GABAergic synapse. This relationship was reliable enough to predict MRS-GABA from NMM-GABA. These findings clarify the mesoscopic underpinnings of GABA levels measured by MRS. Our work will help fulfil the promises of MRS-GABA, enhancing our understanding of human behaviour, brain physiology and pathophysiology. KEY POINTS: GABA neurotransmission is essential for synaptic plasticity and learning (especially motor learning) and is altered in several brain disorders, such as epilepsy and stroke. Quantification of GABA in the human brain is typically obtained by magnetic resonance spectroscopy (MRS). However, the interpretation of MRS-GABA is still debated. By using a biophysical neural mass model, here we show that MRS-GABA relates to physiological measures of tonic inhibition in the human cortex.
Collapse
Affiliation(s)
- Ilenia Paparella
- GIGA-Research, CRC-Human Imaging Unit, 8 allée du Six Août, Batiment B30, University of Liège, Liège, Belgium
| | - Paolo Cardone
- GIGA-Research, Coma Science Group, GIGA-Consciousness, University of Liège, Liège, Belgium
| | - Benedetta Zanichelli
- GIGA-Research, CRC-Human Imaging Unit, 8 allée du Six Août, Batiment B30, University of Liège, Liège, Belgium
| | - Laurent Lamalle
- GIGA-Research, CRC-Human Imaging Unit, 8 allée du Six Août, Batiment B30, University of Liège, Liège, Belgium
| | - Fabienne Collette
- GIGA-Research, CRC-Human Imaging Unit, 8 allée du Six Août, Batiment B30, University of Liège, Liège, Belgium
| | - Siya Sherif
- GIGA-Research, CRC-Human Imaging Unit, 8 allée du Six Août, Batiment B30, University of Liège, Liège, Belgium
| | - Mikhail Zubkov
- GIGA-Research, CRC-Human Imaging Unit, 8 allée du Six Août, Batiment B30, University of Liège, Liège, Belgium
| | - William T Clarke
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Charlotte J Stagg
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Medical Research Council Brain Network Dynamics Unit, University of Oxford, Oxford, UK
| | - Pierre Maquet
- GIGA-Research, CRC-Human Imaging Unit, 8 allée du Six Août, Batiment B30, University of Liège, Liège, Belgium
- Department of Neurology, Domaine Universitaire du Sart Tilman, CHU de Liège, Belgium
| | - Gilles Vandewalle
- GIGA-Research, CRC-Human Imaging Unit, 8 allée du Six Août, Batiment B30, University of Liège, Liège, Belgium
| |
Collapse
|
3
|
Doherty DW, Chen L, Smith Y, Wichmann T, Chu HY, Lytton WW. Decreased cellular excitability of pyramidal tract neurons in primary motor cortex leads to paradoxically increased network activity in simulated parkinsonian motor cortex. RESEARCH SQUARE 2025:rs.3.rs-6254909. [PMID: 40297688 PMCID: PMC12036466 DOI: 10.21203/rs.3.rs-6254909/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Recent evidence suggests that the primary motor cortex (M1) layer 5B pyramidal tract (PT5B) neurons show a decreased intrinsic excitability in mouse models of parkinsonism, which perhaps plays an important role in the pathophysiology of parkinsonian motor symptoms. PT5B neurons project to outputs in the brainstem and the spinal cord, leading to the direct motor expression of Parkinson's disease (PD) pathology. We set out to explore how the decreased PT5B neuron excitability influences the activity patterns of the M1 network. Using NEURON/NetPyNE simulators, we implemented detailed computer simulations of PT5B neurons based on control and 6-OHDA-treated mouse slice data. We placed these PT5B cells in an in vivo M1 network simulation, driven by ascending input from the thalamus and from other cortical areas. Simulated 6-OHDA-treated mouse PT5B neurons in an otherwise unmodified simulated M1 network resulted in major changes in LFP oscillatory power in the parkinsonian condition: an order of magnitude increase in beta band power around 15 Hz in the rest state and a lesser increase in beta power in the parkinsonian activated (movement) state. We demonstrated that relatively small changes in PT5B neuron excitability altered the patterns of activity throughout the M1 circuit. In particular, the decreased PT5B neuron excitability resulted in increased beta band power, which is a signature of PD pathophysiology.
Collapse
|
4
|
Doherty DW, Jung J, Dura-Bernal, Lytton WW. Self-organized and self-sustained ensemble activity patterns in simulation of mouse primary motor cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632866. [PMID: 39868170 PMCID: PMC11760730 DOI: 10.1101/2025.01.13.632866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The idea of self-organized signal processing in the cerebral cortex has become a focus of research since Beggs and Plentz1 reported avalanches in local field potential recordings from organotypic cultures and acute slices of rat somatosensory cortex. How the cortex intrinsically organizes signals remains unknown. A current hypothesis was proposed by the condensed matter physicists Bak, Tang, and Wiesenfeld2 when they conjectured that if neuronal avalanche activity followed inverse power law distributions, then brain activity may be set around phase transitions within self-organized signals. We asked if we would observe self-organized signals in an isolated slice of our data driven detailed simulation of the mouse primary motor cortex? If we did, would we observe avalanches with power-law distributions in size and duration and what would they look like? Our results demonstrate that a brief unstructured stimulus (100ms, 57μA current) to a small subset of neurons (about 181 of more than 10,000) in a simulated mouse primary motor cortex slice results in self-organized and self-sustained avalanches with power-law size and duration distributions and values similar to those reported from in vivo and in vitro experiments. We observed 4 cross-layer and cross-neuron population patterns, 3 of which displayed a dominant rhythmic component. Avalanches were each composed of one or more of the 4 population patterns.
Collapse
Affiliation(s)
- D W Doherty
- Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - J Jung
- Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Dura-Bernal
- Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - W W Lytton
- Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Kings County Hospital, Brooklyn, NY 11203, USA
| |
Collapse
|
5
|
Yu GJ, Ranieri F, Di Lazzaro V, Sommer MA, Peterchev AV, Grill WM. Circuits and mechanisms for TMS-induced corticospinal waves: Connecting sensitivity analysis to the network graph. PLoS Comput Biol 2024; 20:e1012640. [PMID: 39637241 DOI: 10.1371/journal.pcbi.1012640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 12/17/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Transcranial magnetic stimulation (TMS) is a non-invasive, FDA-cleared treatment for neuropsychiatric disorders with broad potential for new applications, but the neural circuits that are engaged during TMS are still poorly understood. Recordings of neural activity from the corticospinal tract provide a direct readout of the response of motor cortex to TMS, and therefore a new opportunity to model neural circuit dynamics. The study goal was to use epidural recordings from the cervical spine of human subjects to develop a computational model of a motor cortical macrocolumn through which the mechanisms underlying the response to TMS, including direct and indirect waves, could be investigated. An in-depth sensitivity analysis was conducted to identify important pathways, and machine learning was used to identify common circuit features among these pathways. Sensitivity analysis identified neuron types that preferentially contributed to single corticospinal waves. Single wave preference could be predicted using the average connection probability of all possible paths between the activated neuron type and L5 pyramidal tract neurons (PTNs). For these activations, the total conduction delay of the shortest path to L5 PTNs determined the latency of the corticospinal wave. Finally, there were multiple neuron type activations that could preferentially modulate a particular corticospinal wave. The results support the hypothesis that different pathways of circuit activation contribute to different corticospinal waves with participation of both excitatory and inhibitory neurons. Moreover, activation of both afferents to the motor cortex as well as specific neuron types within the motor cortex initiated different I-waves, and the results were interpreted to propose the cortical origins of afferents that may give rise to certain I-waves. The methodology provides a workflow for performing computationally tractable sensitivity analyses on complex models and relating the results to the network structure to both identify and understand mechanisms underlying the response to acute stimulation.
Collapse
Affiliation(s)
- Gene J Yu
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, United States of America
| | - Federico Ranieri
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Vincenzo Di Lazzaro
- Department of Medicine and Surgery, Unit of Neurology, Neurophysiology, Neurobiology and Psychiatry, Università Campus Bio-Medico di Roma, Roma, Italy
- Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Marc A Sommer
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Angel V Peterchev
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, United States of America
- Department of Electrical and Computer Engineering, Duke University, Durham, North Carolina, United States of America
- Department of Neurosurgery, Duke University, Durham, North Carolina, United States of America
| | - Warren M Grill
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Department of Electrical and Computer Engineering, Duke University, Durham, North Carolina, United States of America
- Department of Neurosurgery, Duke University, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
6
|
Haghir H, Kuckertz A, Zhao L, Hami J, Palomero-Gallagher N. A new map of the rat isocortex and proisocortex: cytoarchitecture and M 2 receptor distribution patterns. Brain Struct Funct 2024; 229:1795-1822. [PMID: 37318645 PMCID: PMC11485150 DOI: 10.1007/s00429-023-02654-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/13/2023] [Indexed: 06/16/2023]
Abstract
Neurotransmitters and their receptors are key molecules in information transfer between neurons, thus enabling inter-areal communication. Therefore, multimodal atlases integrating the brain's cyto- and receptor architecture constitute crucial tools to understand the relationship between its structural and functional segregation. Cholinergic muscarinic M2 receptors have been shown to be an evolutionarily conserved molecular marker of primary sensory areas in the mammalian brain. To complement existing rodent atlases, we applied a silver cell body staining and quantitative in vitro receptor autoradiographic visualization of M2 receptors to alternating sections throughout the entire brain of five adult male Wistar rats (three sectioned coronally, one horizontally, one sagittally). Histological sections and autoradiographs were scanned at a spatial resolution of 1 µm and 20 µm per pixel, respectively, and files were stored as 8 bit images. We used these high-resolution datasets to create an atlas of the entire rat brain, including the olfactory bulb, cerebellum and brainstem. We describe the cyto- and M2 receptor architectonic features of 48 distinct iso- and proisocortical areas across the rat forebrain and provide their mean M2 receptor density. The ensuing parcellation scheme, which is discussed in the framework of existing comprehensive atlasses, includes the novel subdivision of mediomedial secondary visual area Oc2MM into anterior (Oc2MMa) and posterior (Oc2MMp) parts, and of lateral visual area Oc2L into rostrolateral (Oc2Lr), intermediate dorsolateral (Oc2Lid), intermediate ventrolateral (Oc2Liv) and caudolateral (Oc2Lc) secondary visual areas. The M2 receptor densities and the comprehensive map of iso-and proisocortical areas constitute useful tools for future computational and neuroscientific studies.
Collapse
Affiliation(s)
- Hossein Haghir
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425, Jülich, Germany
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetic Research Center (MGRC), School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Anika Kuckertz
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425, Jülich, Germany
| | - Ling Zhao
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425, Jülich, Germany
| | - Javad Hami
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425, Jülich, Germany
- Faculty of Medicine, HMU Health and Medical University Potsdam, 14471, Potsdam, Germany
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425, Jülich, Germany.
- C. & O. Vogt Institute of Brain Research, Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany.
| |
Collapse
|
7
|
Demchuk AM, Esteves IM, Chang H, Sun J, McNaughton BL. Hierarchical Gradients of Encoded Spatial and Sensory Information in the Neocortex Are Attenuated by Dorsal Hippocampal Lesions. J Neurosci 2024; 44:e1619232024. [PMID: 38942472 PMCID: PMC11293447 DOI: 10.1523/jneurosci.1619-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/16/2024] [Accepted: 05/13/2024] [Indexed: 06/30/2024] Open
Abstract
During navigation, the neocortex actively integrates learned spatial context with current sensory experience to guide behaviors. However, the relative encoding of spatial and sensorimotor information among cortical cells, and whether hippocampal feedback continues to modify these properties after learning, remains poorly understood. Thus, two-photon microscopy of male and female Thy1-GCaMP6s mice was used to longitudinally image neurons spanning superficial retrosplenial cortex and layers II-Va of primary and secondary motor cortices before and after bilateral dorsal hippocampal lesions. During behavior on a familiar cued treadmill, the locations of two obstacles were interchanged to decouple place-tuning from cue-tuning among position-correlated cells with fields at those locations. Subpopulations of place and cue cells each formed interareal gradients such that higher-level cortical regions exhibited higher fractions of place cells, whereas lower-level regions exhibited higher fractions of cue cells. Position-correlated cells in the motor cortex also formed translaminar gradients; more superficial cells were more likely to exhibit fields and were more sparsely and precisely tuned than deeper cells. After dorsal hippocampal lesions, a neural representation of the learned environment persisted, but retrosplenial cortex exhibited significantly increased cue-tuning, and, in motor cortices, both position-correlated cell recruitment and population activity at the unstable obstacle locations became more homogeneously elevated across laminae. Altogether, these results support that the hippocampus continues to modulate cortical responses in familiar environments, and the relative impact of descending feedback obeys hierarchical interareal and interlaminar gradients opposite to the flow of ascending sensory inputs.
Collapse
Affiliation(s)
- Aubrey M Demchuk
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
| | - Ingrid M Esteves
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
| | - HaoRan Chang
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
| | - Jianjun Sun
- Hotchkiss Brain Institute, University of Calgary Foothills, Calgary, Alberta T2N 4N1, Canada
| | - Bruce L McNaughton
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
- Department of Neurobiology and Behaviour, University of California, Irvine, Irvine, California 92697
| |
Collapse
|
8
|
Bhagwandin A, Molnár Z, Bertelsen MF, Karlsson KÆ, Alagaili AN, Bennett NC, Hof PR, Kaswera-Kyamakya C, Gilissen E, Jayakumar J, Manger PR. Where Do Core Thalamocortical Axons Terminate in Mammalian Neocortex When There Is No Cytoarchitecturally Distinct Layer 4? J Comp Neurol 2024; 532:e25652. [PMID: 38962882 DOI: 10.1002/cne.25652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/20/2024] [Accepted: 06/07/2024] [Indexed: 07/05/2024]
Abstract
Although the mammalian cerebral cortex is most often described as a hexalaminar structure, there are cortical areas (primary motor cortex) and species (elephants, cetaceans, and hippopotami), where a cytoarchitecturally indistinct, or absent, layer 4 is noted. Thalamocortical projections from the core, or first order, thalamic system terminate primarily in layers 4/inner 3. We explored the termination sites of core thalamocortical projections in cortical areas and in species where there is no cytoarchitecturally distinct layer 4 using the immunolocalization of vesicular glutamate transporter 2, a known marker of core thalamocortical axon terminals, in 31 mammal species spanning the eutherian radiation. Several variations from the canonical cortical column outline of layer 4 and core thalamocortical inputs were noted. In shrews/microchiropterans, layer 4 was present, but many core thalamocortical projections terminated in layer 1 in addition to layers 4 and inner 3. In primate primary visual cortex, the sublaminated layer 4 was associated with a specialized core thalamocortical projection pattern. In primate primary motor cortex, no cytoarchitecturally distinct layer 4 was evident and the core thalamocortical projections terminated throughout layer 3. In the African elephant, cetaceans, and river hippopotamus, no cytoarchitecturally distinct layer 4 was observed and core thalamocortical projections terminated primarily in inner layer 3 and less densely in outer layer 3. These findings are contextualized in terms of cortical processing, perception, and the evolutionary trajectory leading to an indistinct or absent cortical layer 4.
Collapse
Affiliation(s)
- Adhil Bhagwandin
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford, UK
| | - Mads F Bertelsen
- Centre for Zoo and Wild Animal Health, Copenhagen Zoo, Frederiksberg, Denmark
| | - Karl Æ Karlsson
- Biomedical Engineering, Reykjavik University, Reykjavik, Iceland
| | | | - Nigel C Bennett
- South African Research Chair of Mammal Behavioural Ecology and Physiology, University of Pretoria, Pretoria, South Africa
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Center for Discovery and Innovation, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Emmanuel Gilissen
- Department of African Zoology, Royal Museum for Central Africa, Tervuren, Belgium
- Laboratory of Histology and Neuropathology, Université Libre de Bruxelles, Brussels, Belgium
| | - Jaikishan Jayakumar
- Sudha Gopalakrishnan Brain Centre, Indian Institute of Technology Madras, Chennai, India
- Center for Computational Brain Research, Indian Institute of Technology Madras, Chennai, India
| | - Paul R Manger
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Republic of South Africa
| |
Collapse
|
9
|
Doherty DW, Chen L, Smith Y, Wichmann T, Chu HY, Lytton WW. Decreased cellular excitability of pyramidal tract neurons in primary motor cortex leads to paradoxically increased network activity in simulated parkinsonian motor cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.23.595566. [PMID: 38948850 PMCID: PMC11212883 DOI: 10.1101/2024.05.23.595566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Decreased excitability of pyramidal tract neurons in layer 5B (PT5B) of primary motor cortex (M1) has recently been shown in a dopamine-depleted mouse model of parkinsonism. We hypothesized that decreased PT5B neuron excitability would substantially disrupt oscillatory and non-oscillatory firing patterns of neurons in layer 5 (L5) of primary motor cortex (M1). To test this hypothesis, we performed computer simulations using a previously validated computer model of mouse M1. Inclusion of the experimentally identified parkinsonism-associated decrease of PT5B excitability into our computational model produced a paradoxical increase in rest-state PT5B firing rate, as well as an increase in beta-band oscillatory power in local field potential (LFP). In the movement-state, PT5B population firing and LFP showed reduced beta and increased high-beta, low-gamma activity of 20-35 Hz in the parkinsonian, but not in control condition. The appearance of beta-band oscillations in parkinsonism would be expected to disrupt normal M1 motor output and contribute to motor activity deficits seen in patients with Parkinson's disease (PD).
Collapse
Affiliation(s)
- Donald W Doherty
- Department of Physiology & Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - Liqiang Chen
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington D.C., USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - Yoland Smith
- Emory National Primate Research Center, Department of Neurology, Udall Center of Excellence for Parkinson's Disease Research, Emory University, School of Medicine, Atlanta GA 30329 USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - Thomas Wichmann
- Emory National Primate Research Center, Department of Neurology, Udall Center of Excellence for Parkinson's Disease Research, Emory University, School of Medicine, Atlanta GA 30329 USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - Hong-Yuan Chu
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington D.C., USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - William W Lytton
- Department of Physiology & Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
- Kings County Hospital, Brooklyn, NY 11203, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| |
Collapse
|
10
|
Haggie L, Besier T, McMorland A. Circuits in the motor cortex explain oscillatory responses to transcranial magnetic stimulation. Netw Neurosci 2024; 8:96-118. [PMID: 38562291 PMCID: PMC10861165 DOI: 10.1162/netn_a_00341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/11/2023] [Indexed: 04/04/2024] Open
Abstract
Transcranial magnetic stimulation (TMS) is a popular method used to investigate brain function. Stimulation over the motor cortex evokes muscle contractions known as motor evoked potentials (MEPs) and also high-frequency volleys of electrical activity measured in the cervical spinal cord. The physiological mechanisms of these experimentally derived responses remain unclear, but it is thought that the connections between circuits of excitatory and inhibitory neurons play a vital role. Using a spiking neural network model of the motor cortex, we explained the generation of waves of activity, so called 'I-waves', following cortical stimulation. The model reproduces a number of experimentally known responses including direction of TMS, increased inhibition, and changes in strength. Using populations of thousands of neurons in a model of cortical circuitry we showed that the cortex generated transient oscillatory responses without any tuning, and that neuron parameters such as refractory period and delays influenced the pattern and timing of those oscillations. By comparing our network with simpler, previously proposed circuits, we explored the contributions of specific connections and found that recurrent inhibitory connections are vital in producing later waves that significantly impact the production of motor evoked potentials in downstream muscles (Thickbroom, 2011). This model builds on previous work to increase our understanding of how complex circuitry of the cortex is involved in the generation of I-waves.
Collapse
Affiliation(s)
- Lysea Haggie
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Thor Besier
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Angus McMorland
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Department of Exercise Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
11
|
Luu P, Tucker DM, Friston K. From active affordance to active inference: vertical integration of cognition in the cerebral cortex through dual subcortical control systems. Cereb Cortex 2024; 34:bhad458. [PMID: 38044461 DOI: 10.1093/cercor/bhad458] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 12/05/2023] Open
Abstract
In previous papers, we proposed that the dorsal attention system's top-down control is regulated by the dorsal division of the limbic system, providing a feedforward or impulsive form of control generating expectancies during active inference. In contrast, we proposed that the ventral attention system is regulated by the ventral limbic division, regulating feedback constraints and error-correction for active inference within the neocortical hierarchy. Here, we propose that these forms of cognitive control reflect vertical integration of subcortical arousal control systems that evolved for specific forms of behavior control. The feedforward impetus to action is regulated by phasic arousal, mediated by lemnothalamic projections from the reticular activating system of the lower brainstem, and then elaborated by the hippocampus and dorsal limbic division. In contrast, feedback constraint-based on environmental requirements-is regulated by the tonic activation furnished by collothalamic projections from the midbrain arousal control centers, and then sustained and elaborated by the amygdala, basal ganglia, and ventral limbic division. In an evolutionary-developmental analysis, understanding these differing forms of active affordance-for arousal and motor control within the subcortical vertebrate neuraxis-may help explain the evolution of active inference regulating the cognition of expectancy and error-correction within the mammalian 6-layered neocortex.
Collapse
Affiliation(s)
- Phan Luu
- Brain Electrophysiology Laboratory Company, Riverfront Research Park, 1776 Millrace Dr., Eugene, OR 97403, United States
- Department of Psychology, University of Oregon, Eugene, OR 97403, United States
| | - Don M Tucker
- Brain Electrophysiology Laboratory Company, Riverfront Research Park, 1776 Millrace Dr., Eugene, OR 97403, United States
- Department of Psychology, University of Oregon, Eugene, OR 97403, United States
| | - Karl Friston
- The Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, London WC1N 3AR, United Kingdom
- VERSES AI Research Lab, Los Angeles, CA 90016, USA
| |
Collapse
|
12
|
Nolan M, Scott C, Hof PR, Ansorge O. Betz cells of the primary motor cortex. J Comp Neurol 2024; 532:e25567. [PMID: 38289193 PMCID: PMC10952528 DOI: 10.1002/cne.25567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/11/2023] [Accepted: 11/17/2023] [Indexed: 02/01/2024]
Abstract
Betz cells, named in honor of Volodymyr Betz (1834-1894), who described them as "giant pyramids" in the primary motor cortex of primates and other mammalian species, are layer V extratelencephalic projection (ETP) neurons that directly innervate α-motoneurons of the brainstem and spinal cord. Despite their large volume and circumferential dendritic architecture, to date, no single molecular criterion has been established that unequivocally distinguishes adult Betz cells from other layer V ETP neurons. In primates, transcriptional signatures suggest the presence of at least two ETP neuron clusters that contain mature Betz cells; these are characterized by an abundance of axon guidance and oxidative phosphorylation transcripts. How neurodevelopmental programs drive the distinct positional and morphological features of Betz cells in humans remains unknown. Betz cells display a distinct biphasic firing pattern involving early cessation of firing followed by delayed sustained acceleration in spike frequency and magnitude. Few cell type-specific transcripts and electrophysiological characteristics are conserved between rodent layer V ETP neurons of the motor cortex and primate Betz cells. This has implications for the modeling of disorders that affect the motor cortex in humans, such as amyotrophic lateral sclerosis (ALS). Perhaps vulnerability to ALS is linked to the evolution of neural networks for fine motor control reflected in the distinct morphomolecular architecture of the human motor cortex, including Betz cells. Here, we discuss histological, molecular, and functional data concerning the position of Betz cells in the emerging taxonomy of neurons across diverse species and their role in neurological disorders.
Collapse
Affiliation(s)
- Matthew Nolan
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Connor Scott
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Patrick. R. Hof
- Nash Family Department of Neuroscience and Friedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Olaf Ansorge
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| |
Collapse
|
13
|
Dura-Bernal S, Griffith EY, Barczak A, O'Connell MN, McGinnis T, Moreira JVS, Schroeder CE, Lytton WW, Lakatos P, Neymotin SA. Data-driven multiscale model of macaque auditory thalamocortical circuits reproduces in vivo dynamics. Cell Rep 2023; 42:113378. [PMID: 37925640 PMCID: PMC10727489 DOI: 10.1016/j.celrep.2023.113378] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/05/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023] Open
Abstract
We developed a detailed model of macaque auditory thalamocortical circuits, including primary auditory cortex (A1), medial geniculate body (MGB), and thalamic reticular nucleus, utilizing the NEURON simulator and NetPyNE tool. The A1 model simulates a cortical column with over 12,000 neurons and 25 million synapses, incorporating data on cell-type-specific neuron densities, morphology, and connectivity across six cortical layers. It is reciprocally connected to the MGB thalamus, which includes interneurons and core and matrix-layer-specific projections to A1. The model simulates multiscale measures, including physiological firing rates, local field potentials (LFPs), current source densities (CSDs), and electroencephalography (EEG) signals. Laminar CSD patterns, during spontaneous activity and in response to broadband noise stimulus trains, mirror experimental findings. Physiological oscillations emerge spontaneously across frequency bands comparable to those recorded in vivo. We elucidate population-specific contributions to observed oscillation events and relate them to firing and presynaptic input patterns. The model offers a quantitative theoretical framework to integrate and interpret experimental data and predict its underlying cellular and circuit mechanisms.
Collapse
Affiliation(s)
- Salvador Dura-Bernal
- Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY, USA; Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| | - Erica Y Griffith
- Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY, USA; Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| | - Annamaria Barczak
- Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Monica N O'Connell
- Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Tammy McGinnis
- Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Joao V S Moreira
- Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY, USA
| | - Charles E Schroeder
- Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Departments of Psychiatry and Neurology, Columbia University Medical Center, New York, NY, USA
| | - William W Lytton
- Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY, USA; Kings County Hospital Center, Brooklyn, NY, USA
| | - Peter Lakatos
- Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Samuel A Neymotin
- Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department Psychiatry, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Kogan E, Lu J, Zuo Y. Cortical circuit dynamics underlying motor skill learning: from rodents to humans. Front Mol Neurosci 2023; 16:1292685. [PMID: 37965043 PMCID: PMC10641381 DOI: 10.3389/fnmol.2023.1292685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Motor learning is crucial for the survival of many animals. Acquiring a new motor skill involves complex alterations in both local neural circuits in many brain regions and long-range connections between them. Such changes can be observed anatomically and functionally. The primary motor cortex (M1) integrates information from diverse brain regions and plays a pivotal role in the acquisition and refinement of new motor skills. In this review, we discuss how motor learning affects the M1 at synaptic, cellular, and circuit levels. Wherever applicable, we attempt to relate and compare findings in humans, non-human primates, and rodents. Understanding the underlying principles shared by different species will deepen our understanding of the neurobiological and computational basis of motor learning.
Collapse
Affiliation(s)
| | | | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
15
|
Jorstad NL, Close J, Johansen N, Yanny AM, Barkan ER, Travaglini KJ, Bertagnolli D, Campos J, Casper T, Crichton K, Dee N, Ding SL, Gelfand E, Goldy J, Hirschstein D, Kroll M, Kunst M, Lathia K, Long B, Martin N, McMillen D, Pham T, Rimorin C, Ruiz A, Shapovalova N, Shehata S, Siletti K, Somasundaram S, Sulc J, Tieu M, Torkelson A, Tung H, Ward K, Callaway EM, Hof PR, Keene CD, Levi BP, Linnarsson S, Mitra PP, Smith K, Hodge RD, Bakken TE, Lein ES. Transcriptomic cytoarchitecture reveals principles of human neocortex organization. Science 2023; 382:eadf6812. [PMID: 37824655 PMCID: PMC11687949 DOI: 10.1126/science.adf6812] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 09/08/2023] [Indexed: 10/14/2023]
Abstract
Variation in cytoarchitecture is the basis for the histological definition of cortical areas. We used single cell transcriptomics and performed cellular characterization of the human cortex to better understand cortical areal specialization. Single-nucleus RNA-sequencing of 8 areas spanning cortical structural variation showed a highly consistent cellular makeup for 24 cell subclasses. However, proportions of excitatory neuron subclasses varied substantially, likely reflecting differences in connectivity across primary sensorimotor and association cortices. Laminar organization of astrocytes and oligodendrocytes also differed across areas. Primary visual cortex showed characteristic organization with major changes in the excitatory to inhibitory neuron ratio, expansion of layer 4 excitatory neurons, and specialized inhibitory neurons. These results lay the groundwork for a refined cellular and molecular characterization of human cortical cytoarchitecture and areal specialization.
Collapse
Affiliation(s)
| | - Jennie Close
- Allen Institute for Brain Science; Seattle, WA, 98109
| | | | | | | | | | | | - Jazmin Campos
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Tamara Casper
- Allen Institute for Brain Science; Seattle, WA, 98109
| | | | - Nick Dee
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Song-Lin Ding
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Emily Gelfand
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Jeff Goldy
- Allen Institute for Brain Science; Seattle, WA, 98109
| | | | - Matthew Kroll
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Michael Kunst
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Kanan Lathia
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Brian Long
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Naomi Martin
- Allen Institute for Brain Science; Seattle, WA, 98109
| | | | | | | | - Augustin Ruiz
- Allen Institute for Brain Science; Seattle, WA, 98109
| | | | | | - Kimberly Siletti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet; Stockholm, Sweden, 171 77
| | | | - Josef Sulc
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Michael Tieu
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Amy Torkelson
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Herman Tung
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Katelyn Ward
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Edward M. Callaway
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, 92037
| | - Patrick R. Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, WA, 98195
| | - Boaz P. Levi
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Sten Linnarsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet; Stockholm, Sweden, 171 77
| | - Partha P. Mitra
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724
| | | | | | | | - Ed S. Lein
- Allen Institute for Brain Science; Seattle, WA, 98109
| |
Collapse
|
16
|
Shi H, He Y, Zhou Y, Huang J, Maher K, Wang B, Tang Z, Luo S, Tan P, Wu M, Lin Z, Ren J, Thapa Y, Tang X, Chan KY, Deverman BE, Shen H, Liu A, Liu J, Wang X. Spatial atlas of the mouse central nervous system at molecular resolution. Nature 2023; 622:552-561. [PMID: 37758947 PMCID: PMC10709140 DOI: 10.1038/s41586-023-06569-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/22/2023] [Indexed: 09/29/2023]
Abstract
Spatially charting molecular cell types at single-cell resolution across the 3D volume is critical for illustrating the molecular basis of brain anatomy and functions. Single-cell RNA sequencing has profiled molecular cell types in the mouse brain1,2, but cannot capture their spatial organization. Here we used an in situ sequencing method, STARmap PLUS3,4, to profile 1,022 genes in 3D at a voxel size of 194 × 194 × 345 nm3, mapping 1.09 million high-quality cells across the adult mouse brain and spinal cord. We developed computational pipelines to segment, cluster and annotate 230 molecular cell types by single-cell gene expression and 106 molecular tissue regions by spatial niche gene expression. Joint analysis of molecular cell types and molecular tissue regions enabled a systematic molecular spatial cell-type nomenclature and identification of tissue architectures that were undefined in established brain anatomy. To create a transcriptome-wide spatial atlas, we integrated STARmap PLUS measurements with a published single-cell RNA-sequencing atlas1, imputing single-cell expression profiles of 11,844 genes. Finally, we delineated viral tropisms of a brain-wide transgene delivery tool, AAV-PHP.eB5,6. Together, this annotated dataset provides a single-cell resource that integrates the molecular spatial atlas, brain anatomy and the accessibility to genetic manipulation of the mammalian central nervous system.
Collapse
Affiliation(s)
- Hailing Shi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yichun He
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Yiming Zhou
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jiahao Huang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kamal Maher
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computational and Systems Biology PhD Program, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brandon Wang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zefang Tang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shuchen Luo
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peng Tan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Morgan Wu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zuwan Lin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Jingyi Ren
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yaman Thapa
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xin Tang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Ken Y Chan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Benjamin E Deverman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hao Shen
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Albert Liu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA.
| | - Xiao Wang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
17
|
Matsuda K, Shirakami A, Nakajima R, Akutsu T, Shimono M. Whole-Brain Evaluation of Cortical Microconnectomes. eNeuro 2023; 10:ENEURO.0094-23.2023. [PMID: 37903612 PMCID: PMC10616907 DOI: 10.1523/eneuro.0094-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/08/2023] [Accepted: 09/30/2023] [Indexed: 11/01/2023] Open
Abstract
The brain is an organ that functions as a network of many elements connected in a nonuniform manner. In the brain, the neocortex is evolutionarily newest and is thought to be primarily responsible for the high intelligence of mammals. In the mature mammalian brain, all cortical regions are expected to have some degree of homology, but have some variations of local circuits to achieve specific functions performed by individual regions. However, few cellular-level studies have examined how the networks within different cortical regions differ. This study aimed to find rules for systematic changes of connectivity (microconnectomes) across 16 different cortical region groups. We also observed unknown trends in basic parameters in vitro such as firing rate and layer thickness across brain regions. Results revealed that the frontal group shows unique characteristics such as dense active neurons, thick cortex, and strong connections with deeper layers. This suggests the frontal side of the cortex is inherently capable of driving, even in isolation and that frontal nodes provide the driving force generating a global pattern of spontaneous synchronous activity, such as the default mode network. This finding provides a new hypothesis explaining why disruption in the frontal region causes a large impact on mental health.
Collapse
Affiliation(s)
- Kouki Matsuda
- Graduate Schools of Medicine, Kyoto University, 53 Kawaramachi, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Arata Shirakami
- Graduate Schools of Medicine, Kyoto University, 53 Kawaramachi, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ryota Nakajima
- Graduate Schools of Medicine, Kyoto University, 53 Kawaramachi, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
| | - Masanori Shimono
- Graduate Schools of Medicine, Kyoto University, 53 Kawaramachi, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
- Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita-shi, Osaka 565-0871
| |
Collapse
|
18
|
Dura-Bernal S, Neymotin SA, Suter BA, Dacre J, Moreira JVS, Urdapilleta E, Schiemann J, Duguid I, Shepherd GMG, Lytton WW. Multiscale model of primary motor cortex circuits predicts in vivo cell-type-specific, behavioral state-dependent dynamics. Cell Rep 2023; 42:112574. [PMID: 37300831 PMCID: PMC10592234 DOI: 10.1016/j.celrep.2023.112574] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 02/27/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023] Open
Abstract
Understanding cortical function requires studying multiple scales: molecular, cellular, circuit, and behavioral. We develop a multiscale, biophysically detailed model of mouse primary motor cortex (M1) with over 10,000 neurons and 30 million synapses. Neuron types, densities, spatial distributions, morphologies, biophysics, connectivity, and dendritic synapse locations are constrained by experimental data. The model includes long-range inputs from seven thalamic and cortical regions and noradrenergic inputs. Connectivity depends on cell class and cortical depth at sublaminar resolution. The model accurately predicts in vivo layer- and cell-type-specific responses (firing rates and LFP) associated with behavioral states (quiet wakefulness and movement) and experimental manipulations (noradrenaline receptor blockade and thalamus inactivation). We generate mechanistic hypotheses underlying the observed activity and analyzed low-dimensional population latent dynamics. This quantitative theoretical framework can be used to integrate and interpret M1 experimental data and sheds light on the cell-type-specific multiscale dynamics associated with several experimental conditions and behaviors.
Collapse
Affiliation(s)
- Salvador Dura-Bernal
- Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY, USA; Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| | - Samuel A Neymotin
- Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department of Psychiatry, Grossman School of Medicine, New York University (NYU), New York, NY, USA
| | - Benjamin A Suter
- Department of Physiology, Northwestern University, Evanston, IL, USA
| | - Joshua Dacre
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Joao V S Moreira
- Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY, USA
| | - Eugenio Urdapilleta
- Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY, USA
| | - Julia Schiemann
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK; Center for Integrative Physiology and Molecular Medicine, Saarland University, Saarbrücken, Germany
| | - Ian Duguid
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | | | - William W Lytton
- Department of Physiology and Pharmacology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; Department of Neurology, Kings County Hospital Center, Brooklyn, NY, USA
| |
Collapse
|
19
|
Haviv D, Gatie M, Hadjantonakis AK, Nawy T, Pe’er D. The covariance environment defines cellular niches for spatial inference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537375. [PMID: 37131616 PMCID: PMC10153165 DOI: 10.1101/2023.04.18.537375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The tsunami of new multiplexed spatial profiling technologies has opened a range of computational challenges focused on leveraging these powerful data for biological discovery. A key challenge underlying computation is a suitable representation for features of cellular niches. Here, we develop the covariance environment (COVET), a representation that can capture the rich, continuous multivariate nature of cellular niches by capturing the gene-gene covariate structure across cells in the niche, which can reflect the cell-cell communication between them. We define a principled optimal transport-based distance metric between COVET niches and develop a computationally efficient approximation to this metric that can scale to millions of cells. Using COVET to encode spatial context, we develop environmental variational inference (ENVI), a conditional variational autoencoder that jointly embeds spatial and single-cell RNA-seq data into a latent space. Two distinct decoders either impute gene expression across spatial modality, or project spatial information onto dissociated single-cell data. We show that ENVI is not only superior in the imputation of gene expression but is also able to infer spatial context to disassociated single-cell genomics data.
Collapse
Affiliation(s)
- Doron Haviv
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Medicine; New York, NY 10065, USA
| | - Mohamed Gatie
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tal Nawy
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dana Pe’er
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| |
Collapse
|
20
|
West TO, Duchet B, Farmer SF, Friston KJ, Cagnan H. When do bursts matter in the primary motor cortex? Investigating changes in the intermittencies of beta rhythms associated with movement states. Prog Neurobiol 2023; 221:102397. [PMID: 36565984 PMCID: PMC7614511 DOI: 10.1016/j.pneurobio.2022.102397] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/04/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Brain activity exhibits significant temporal structure that is not well captured in the power spectrum. Recently, attention has shifted to characterising the properties of intermittencies in rhythmic neural activity (i.e. bursts), yet the mechanisms that regulate them are unknown. Here, we present evidence from electrocorticography recordings made over the motor cortex to show that the statistics of bursts, such as duration or amplitude, in the beta frequency (14-30 Hz) band, significantly aid the classification of motor states such as rest, movement preparation, execution, and imagery. These features reflect nonlinearities not detectable in the power spectrum, with states increasing in nonlinearity from movement execution to preparation to rest. Further, we show using a computational model of the cortical microcircuit, constrained to account for burst features, that modulations of laminar specific inhibitory interneurons are responsible for the temporal organisation of activity. Finally, we show that the temporal characteristics of spontaneous activity can be used to infer the balance of cortical integration between incoming sensory information and endogenous activity. Critically, we contribute to the understanding of how transient brain rhythms may underwrite cortical processing, which in turn, could inform novel approaches for brain state classification, and modulation with novel brain-computer interfaces.
Collapse
Affiliation(s)
- Timothy O West
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Mansfield Road, Oxford OX1 3TH, UK; Wellcome Centre for Human Neuroimaging, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK.
| | - Benoit Duchet
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Mansfield Road, Oxford OX1 3TH, UK
| | - Simon F Farmer
- Department of Neurology, National Hospital for Neurology & Neurosurgery, Queen Square, London WC1N 3BG, UK; Department of Clinical and Movement Neurosciences, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Karl J Friston
- Wellcome Centre for Human Neuroimaging, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Hayriye Cagnan
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Mansfield Road, Oxford OX1 3TH, UK; Wellcome Centre for Human Neuroimaging, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| |
Collapse
|
21
|
Xu Z, Geron E, Pérez-Cuesta LM, Bai Y, Gan WB. Generalized extinction of fear memory depends on co-allocation of synaptic plasticity in dendrites. Nat Commun 2023; 14:503. [PMID: 36720872 PMCID: PMC9889816 DOI: 10.1038/s41467-023-35805-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/03/2023] [Indexed: 02/02/2023] Open
Abstract
Memories can be modified by new experience in a specific or generalized manner. Changes in synaptic connections are crucial for memory storage, but it remains unknown how synaptic changes associated with different memories are distributed within neuronal circuits and how such distributions affect specific or generalized modification by novel experience. Here we show that fear conditioning with two different auditory stimuli (CS) and footshocks (US) induces dendritic spine elimination mainly on different dendritic branches of layer 5 pyramidal neurons in the mouse motor cortex. Subsequent fear extinction causes CS-specific spine formation and extinction of freezing behavior. In contrast, spine elimination induced by fear conditioning with >2 different CS-USs often co-exists on the same dendritic branches. Fear extinction induces CS-nonspecific spine formation and generalized fear extinction. Moreover, activation of somatostatin-expressing interneurons increases the occurrence of spine elimination induced by different CS-USs on the same dendritic branches and facilitates the generalization of fear extinction. These findings suggest that specific or generalized modification of existing memories by new experience depends on whether synaptic changes induced by previous experiences are segregated or co-exist at the level of individual dendritic branches.
Collapse
Affiliation(s)
- Zhiwei Xu
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Erez Geron
- Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Luis M Pérez-Cuesta
- Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Yang Bai
- Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Wen-Biao Gan
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
- Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| |
Collapse
|
22
|
Abstract
Dendritic spine features in human neurons follow the up-to-date knowledge presented in the previous chapters of this book. Human dendrites are notable for their heterogeneity in branching patterns and spatial distribution. These data relate to circuits and specialized functions. Spines enhance neuronal connectivity, modulate and integrate synaptic inputs, and provide additional plastic functions to microcircuits and large-scale networks. Spines present a continuum of shapes and sizes, whose number and distribution along the dendritic length are diverse in neurons and different areas. Indeed, human neurons vary from aspiny or "relatively aspiny" cells to neurons covered with a high density of intermingled pleomorphic spines on very long dendrites. In this chapter, we discuss the phylogenetic and ontogenetic development of human spines and describe the heterogeneous features of human spiny neurons along the spinal cord, brainstem, cerebellum, thalamus, basal ganglia, amygdala, hippocampal regions, and neocortical areas. Three-dimensional reconstructions of Golgi-impregnated dendritic spines and data from fluorescence microscopy are reviewed with ultrastructural findings to address the complex possibilities for synaptic processing and integration in humans. Pathological changes are also presented, for example, in Alzheimer's disease and schizophrenia. Basic morphological data can be linked to current techniques, and perspectives in this research field include the characterization of spines in human neurons with specific transcriptome features, molecular classification of cellular diversity, and electrophysiological identification of coexisting subpopulations of cells. These data would enlighten how cellular attributes determine neuron type-specific connectivity and brain wiring for our diverse aptitudes and behavior.
Collapse
Affiliation(s)
- Josué Renner
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Alberto A Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
23
|
Kahn M, Krone LB, Blanco‐Duque C, Guillaumin MCC, Mann EO, Vyazovskiy VV. Neuronal-spiking-based closed-loop stimulation during cortical ON- and OFF-states in freely moving mice. J Sleep Res 2022; 31:e13603. [PMID: 35665551 PMCID: PMC9786831 DOI: 10.1111/jsr.13603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/20/2022] [Accepted: 03/22/2022] [Indexed: 12/30/2022]
Abstract
The slow oscillation is a central neuronal dynamic during sleep, and is generated by alternating periods of high and low neuronal activity (ON- and OFF-states). Mounting evidence causally links the slow oscillation to sleep's functions, and it has recently become possible to manipulate the slow oscillation non-invasively and phase-specifically. These developments represent promising clinical avenues, but they also highlight the importance of improving our understanding of how ON/OFF-states affect incoming stimuli and what role they play in neuronal plasticity. Most studies using closed-loop stimulation rely on the electroencephalogram and local field potential signals, which reflect neuronal ON- and OFF-states only indirectly. Here we develop an online detection algorithm based on spiking activity recorded from laminar arrays in mouse motor cortex. We find that online detection of ON- and OFF-states reflects specific phases of spontaneous local field potential slow oscillation. Our neuronal-spiking-based closed-loop procedure offers a novel opportunity for testing the functional role of slow oscillation in sleep-related restorative processes and neural plasticity.
Collapse
Affiliation(s)
- Martin Kahn
- Department of PhysiologyAnatomy and Genetics, University of OxfordOxfordUK,Sleep and Circadian Neuroscience InstituteUniversity of OxfordOxfordUK
| | - Lukas B. Krone
- Department of PhysiologyAnatomy and Genetics, University of OxfordOxfordUK,Sleep and Circadian Neuroscience InstituteUniversity of OxfordOxfordUK,University Hospital of Psychiatry and PsychotherapyUniversity of BernBernSwitzerland,Centre for Experimental NeurologyUniversity of BernBernSwitzerland
| | - Cristina Blanco‐Duque
- Department of PhysiologyAnatomy and Genetics, University of OxfordOxfordUK,Sleep and Circadian Neuroscience InstituteUniversity of OxfordOxfordUK
| | - Mathilde C. C. Guillaumin
- Sleep and Circadian Neuroscience InstituteUniversity of OxfordOxfordUK,Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK,Department of Health Sciences and TechnologyInstitute for NeuroscienceETH, ZurichSwitzerland
| | - Edward O. Mann
- Department of PhysiologyAnatomy and Genetics, University of OxfordOxfordUK
| | - Vladyslav V. Vyazovskiy
- Department of PhysiologyAnatomy and Genetics, University of OxfordOxfordUK,Sleep and Circadian Neuroscience InstituteUniversity of OxfordOxfordUK
| |
Collapse
|
24
|
Chan RW, Cron GO, Asaad M, Edelman BJ, Lee HJ, Adesnik H, Feinberg D, Lee JH. Distinct local and brain-wide networks are activated by optogenetic stimulation of neurons specific to each layer of motor cortex. Neuroimage 2022; 263:119640. [PMID: 36176220 PMCID: PMC10025169 DOI: 10.1016/j.neuroimage.2022.119640] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 11/20/2022] Open
Abstract
Primary motor cortex (M1) consists of a stack of interconnected but distinct layers (L1-L6) which affect motor control through large-scale networks. However, the brain-wide functional influence of each layer is poorly understood. We sought to expand our knowledge of these layers' circuitry by combining Cre-driver mouse lines, optogenetics, fMRI, and electrophysiology. Neuronal activities initiated in Drd3 neurons (within L2/3) were mainly confined within M1, while stimulation of Scnn1a, Rbp4, and Ntsr1 neurons (within L4, L5, and L6, respectively) evoked distinct responses in M1 and motor-related subcortical regions, including striatum and motor thalamus. We also found that fMRI responses from targeted stimulations correlated with both local field potentials (LFPs) and spike changes. This study represents a step forward in our understanding of how different layers of primary motor cortex are embedded in brain-wide circuitry.
Collapse
Affiliation(s)
- Russell W Chan
- Department of Neurology and Neurological Sciences, Stanford University, CA 94305, USA
| | - Greg O Cron
- Department of Neurology and Neurological Sciences, Stanford University, CA 94305, USA
| | - Mazen Asaad
- Department of Molecular and Cellular Physiology, Stanford University, CA 94305, USA
| | - Bradley J Edelman
- Department of Neurology and Neurological Sciences, Stanford University, CA 94305, USA
| | - Hyun Joo Lee
- Department of Neurology and Neurological Sciences, Stanford University, CA 94305, USA
| | - Hillel Adesnik
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | - David Feinberg
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, CA 94305, USA; Department of Bioengineering, Stanford University, CA 94305, USA; Department of Neurosurgery, Stanford University, CA 94305, USA; Department of Electrical Engineering, Stanford University, CA 94305, USA.
| |
Collapse
|
25
|
Nano PR, Bhaduri A. Evaluation of advances in cortical development using model systems. Dev Neurobiol 2022; 82:408-427. [PMID: 35644985 PMCID: PMC10924780 DOI: 10.1002/dneu.22879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 11/11/2022]
Abstract
Compared with that of even the closest primates, the human cortex displays a high degree of specialization and expansion that largely emerges developmentally. Although decades of research in the mouse and other model systems has revealed core tenets of cortical development that are well preserved across mammalian species, small deviations in transcription factor expression, novel cell types in primates and/or humans, and unique cortical architecture distinguish the human cortex. Importantly, many of the genes and signaling pathways thought to drive human-specific cortical expansion also leave the brain vulnerable to disease, as the misregulation of these factors is highly correlated with neurodevelopmental and neuropsychiatric disorders. However, creating a comprehensive understanding of human-specific cognition and disease remains challenging. Here, we review key stages of cortical development and highlight known or possible differences between model systems and the developing human brain. By identifying the developmental trajectories that may facilitate uniquely human traits, we highlight open questions in need of approaches to examine these processes in a human context and reveal translatable insights into human developmental disorders.
Collapse
Affiliation(s)
- Patricia R Nano
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
26
|
Imam A, Bhagwandin A, Ajao MS, Manger PR. The brain of the tree pangolin (Manis tricuspis). IX. The pallial telencephalon. J Comp Neurol 2022; 530:2645-2691. [PMID: 35621013 PMCID: PMC9546464 DOI: 10.1002/cne.25349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/13/2022] [Accepted: 05/04/2022] [Indexed: 11/25/2022]
Abstract
A cyto‐, myelo‐, and chemoarchitectonic analysis of the pallial telencephalon of the tree pangolin is provided. As certain portions of the pallial telencephalon have been described previously (olfactory pallium, hippocampal formation, and amygdaloid complex), we focus on the claustrum and endopiriform nuclear complex, the white matter and white matter interstitial cells, and the areal organization of the cerebral cortex. Our analysis indicates that the organization of the pallial telencephalon of the tree pangolin is similar to that observed in many other mammals, and specifically quite similar to the closely related carnivores. The claustrum of the tree pangolin exhibits a combination of insular and laminar architecture, while the endopiriform nuclear complex contains three nuclei, both reminiscent of observations made in other mammals. The population of white matter interstitial cells resembles that observed in other mammals, while a distinct laminated organization of the intracortical white matter was revealed with parvalbumin immunostaining. The cerebral cortex of the tree pangolin presented with indistinct laminar boundaries as well as pyramidalization of the neurons in both layers 2 and 4. All cortical regions typically found in mammals were present, with the cortical areas within these regions often corresponding to what has been reported in carnivores. Given the similarity of the organization of the pallial telencephalon of the tree pangolin to that observed in other mammals, especially carnivores, it would be reasonable to assume that the neural processing afforded the tree pangolin by these structures does not differ dramatically to that of other mammals.
Collapse
Affiliation(s)
- Aminu Imam
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Republic of South Africa.,Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Adhil Bhagwandin
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Republic of South Africa
| | - Moyosore S Ajao
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Paul R Manger
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Republic of South Africa
| |
Collapse
|
27
|
Tian D, Izumi SI. Transcranial Magnetic Stimulation and Neocortical Neurons: The Micro-Macro Connection. Front Neurosci 2022; 16:866245. [PMID: 35495053 PMCID: PMC9039343 DOI: 10.3389/fnins.2022.866245] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/28/2022] [Indexed: 12/20/2022] Open
Abstract
Understanding the operation of cortical circuits is an important and necessary task in both neuroscience and neurorehabilitation. The functioning of the neocortex results from integrative neuronal activity, which can be probed non-invasively by transcranial magnetic stimulation (TMS). Despite a clear indication of the direct involvement of cortical neurons in TMS, no explicit connection model has been made between the microscopic neuronal landscape and the macroscopic TMS outcome. Here we have performed an integrative review of multidisciplinary evidence regarding motor cortex neurocytology and TMS-related neurophysiology with the aim of elucidating the micro–macro connections underlying TMS. Neurocytological evidence from animal and human studies has been reviewed to describe the landscape of the cortical neurons covering the taxonomy, morphology, circuit wiring, and excitatory–inhibitory balance. Evidence from TMS studies in healthy humans is discussed, with emphasis on the TMS pulse and paradigm selectivity that reflect the underlying neural circuitry constitution. As a result, we propose a preliminary neuronal model of the human motor cortex and then link the TMS mechanisms with the neuronal model by stimulus intensity, direction of induced current, and paired-pulse timing. As TMS bears great developmental potential for both a probe and modulator of neural network activity and neurotransmission, the connection model will act as a foundation for future combined studies of neurocytology and neurophysiology, as well as the technical advances and application of TMS.
Collapse
Affiliation(s)
- Dongting Tian
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduates School of Medicine, Sendai, Japan
- *Correspondence: Dongting Tian,
| | - Shin-Ichi Izumi
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduates School of Medicine, Sendai, Japan
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
- Shin-Ichi Izumi,
| |
Collapse
|
28
|
Kuang H, Liu T, Jiao C, Wang J, Wu S, Wu J, Peng S, Davidson AM, Zeng SX, Lu H, Mostany R. Genetic Deficiency of p53 Leads to Structural, Functional, and Synaptic Deficits in Primary Somatosensory Cortical Neurons of Adult Mice. Front Mol Neurosci 2022; 15:871974. [PMID: 35465090 PMCID: PMC9021533 DOI: 10.3389/fnmol.2022.871974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
The tumor suppressor p53 plays a crucial role in embryonic neuron development and neurite growth, and its involvement in neuronal homeostasis has been proposed. To better understand how the lack of the p53 gene function affects neuronal activity, spine development, and plasticity, we examined the electrophysiological and morphological properties of layer 5 (L5) pyramidal neurons in the primary somatosensory cortex barrel field (S1BF) by using in vitro whole-cell patch clamp and in vivo two-photon imaging techniques in p53 knockout (KO) mice. We found that the spiking frequency, excitatory inputs, and sag ratio were decreased in L5 pyramidal neurons of p53KO mice. In addition, both in vitro and in vivo morphological analyses demonstrated that dendritic spine density in the apical tuft is decreased in L5 pyramidal neurons of p53KO mice. Furthermore, chronic imaging showed that p53 deletion decreased dendritic spine turnover in steady-state conditions, and prevented the increase in spine turnover associated with whisker stimulation seen in wildtype mice. In addition, the sensitivity of whisker-dependent texture discrimination was impaired in p53KO mice compared with wildtype controls. Together, these results suggest that p53 plays an important role in regulating synaptic plasticity by reducing neuronal excitability and the number of excitatory synapses in S1BF.
Collapse
Affiliation(s)
- Haixia Kuang
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Liu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, United States
- *Correspondence: Tao Liu Hua Lu Ricardo Mostany
| | - Cui Jiao
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianmei Wang
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shinan Wu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Wu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Sicong Peng
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Andrew M. Davidson
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Shelya X. Zeng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Hua Lu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, United States
- *Correspondence: Tao Liu Hua Lu Ricardo Mostany
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States
- *Correspondence: Tao Liu Hua Lu Ricardo Mostany
| |
Collapse
|
29
|
Domínguez-Sala E, Valdés-Sánchez L, Canals S, Reiner O, Pombero A, García-López R, Estirado A, Pastor D, Geijo-Barrientos E, Martínez S. Abnormalities in Cortical GABAergic Interneurons of the Primary Motor Cortex Caused by Lis1 (Pafah1b1) Mutation Produce a Non-drastic Functional Phenotype. Front Cell Dev Biol 2022; 10:769853. [PMID: 35309904 PMCID: PMC8924048 DOI: 10.3389/fcell.2022.769853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/31/2022] [Indexed: 11/25/2022] Open
Abstract
LIS1 (PAFAH1B1) plays a major role in the developing cerebral cortex, and haploinsufficient mutations cause human lissencephaly type 1. We have studied morphological and functional properties of the cerebral cortex of mutant mice harboring a deletion in the first exon of the mouse Lis1 (Pafah1b1) gene, which encodes for the LisH domain. The Lis1/sLis1 animals had an overall unaltered cortical structure but showed an abnormal distribution of cortical GABAergic interneurons (those expressing calbindin, calretinin, or parvalbumin), which mainly accumulated in the deep neocortical layers. Interestingly, the study of the oscillatory activity revealed an apparent inability of the cortical circuits to produce correct activity patterns. Moreover, the fast spiking (FS) inhibitory GABAergic interneurons exhibited several abnormalities regarding the size of the action potentials, the threshold for spike firing, the time course of the action potential after-hyperpolarization (AHP), the firing frequency, and the frequency and peak amplitude of spontaneous excitatory postsynaptic currents (sEPSC’s). These morphological and functional alterations in the cortical inhibitory system characterize the Lis1/sLis1 mouse as a model of mild lissencephaly, showing a phenotype less drastic than the typical phenotype attributed to classical lissencephaly. Therefore, the results described in the present manuscript corroborate the idea that mutations in some regions of the Lis1 gene can produce phenotypes more similar to those typically described in schizophrenic and autistic patients and animal models.
Collapse
Affiliation(s)
- E Domínguez-Sala
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - L Valdés-Sánchez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - S Canals
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - O Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - A Pombero
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - R García-López
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - A Estirado
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - D Pastor
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - E Geijo-Barrientos
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain
| | - S Martínez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Sant Joan d'Alacant, Spain.,Centro de Investigación Biomédica en Red en Salud Mental CIBERSAM, Madrid, Spain
| |
Collapse
|
30
|
Harb K, Richter M, Neelagandan N, Magrinelli E, Harfoush H, Kuechler K, Henis M, Hermanns-Borgmeyer I, Calderon de Anda F, Duncan K. Pum2 and TDP-43 refine area-specific cytoarchitecture post-mitotically and modulate translation of Sox5, Bcl11b, and Rorb mRNAs in developing mouse neocortex. eLife 2022; 11:55199. [PMID: 35262486 PMCID: PMC8906809 DOI: 10.7554/elife.55199] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/25/2022] [Indexed: 12/15/2022] Open
Abstract
In the neocortex, functionally distinct areas process specific types of information. Area identity is established by morphogens and transcriptional master regulators, but downstream mechanisms driving area-specific neuronal specification remain unclear. Here, we reveal a role for RNA-binding proteins in defining area-specific cytoarchitecture. Mice lacking Pum2 or overexpressing human TDP-43 show apparent ‘motorization’ of layers IV and V of primary somatosensory cortex (S1), characterized by dramatic expansion of cells co-expressing Sox5 and Bcl11b/Ctip2, a hallmark of subcerebral projection neurons, at the expense of cells expressing the layer IV neuronal marker Rorβ. Moreover, retrograde labeling experiments with cholera toxin B in Pum2; Emx1-Cre and TDP43A315T mice revealed a corresponding increase in subcerebral connectivity of these neurons in S1. Intriguingly, other key features of somatosensory area identity are largely preserved, suggesting that Pum2 and TDP-43 may function in a downstream program, rather than controlling area identity per se. Transfection of primary neurons and in utero electroporation (IUE) suggest cell-autonomous and post-mitotic modulation of Sox5, Bcl11b/Ctip2, and Rorβ levels. Mechanistically, we find that Pum2 and TDP-43 directly interact with and affect the translation of mRNAs encoding Sox5, Bcl11b/Ctip2, and Rorβ. In contrast, effects on the levels of these mRNAs were not detectable in qRT-PCR or single-molecule fluorescent in situ hybridization assays, and we also did not detect effects on their splicing or polyadenylation patterns. Our results support the notion that post-transcriptional regulatory programs involving translational regulation and mediated by Pum2 and TDP-43 contribute to elaboration of area-specific neuronal identity and connectivity in the neocortex.
Collapse
Affiliation(s)
- Kawssar Harb
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Melanie Richter
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nagammal Neelagandan
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Elia Magrinelli
- Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | - Hend Harfoush
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Katrin Kuechler
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Melad Henis
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Anatomy and Histology, Faculty of Veterinary Medicine, New Valley University, New Valley, Egypt
| | - Irm Hermanns-Borgmeyer
- Transgenic Service Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Froylan Calderon de Anda
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kent Duncan
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
31
|
Cousineau J, Plateau V, Baufreton J, Le Bon-Jégo M. Dopaminergic modulation of primary motor cortex: From cellular and synaptic mechanisms underlying motor learning to cognitive symptoms in Parkinson's disease. Neurobiol Dis 2022; 167:105674. [PMID: 35245676 DOI: 10.1016/j.nbd.2022.105674] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 11/16/2022] Open
Abstract
The primary motor cortex (M1) is crucial for movement execution, especially dexterous ones, but also for cognitive functions like motor learning. The acquisition of motor skills to execute dexterous movements requires dopamine-dependent and -independent plasticity mechanisms within M1. In addition to the basal ganglia, M1 is disturbed in Parkinson's disease (PD). However, little is known about how the lack of dopamine (DA), characteristic of PD, directly or indirectly impacts M1 circuitry. Here we review data from studies of PD patients and the substantial research in non-human primate and rodent models of DA depletion. These models enable us to understand the importance of DA in M1 physiology at the behavioral, network, cellular, and synaptic levels. We first summarize M1 functions and neuronal populations in mammals. We then look at the origin of M1 DA and the cellular location of its receptors and explore the impact of DA loss on M1 physiology, motor, and executive functions. Finally, we discuss how PD treatments impact M1 functions.
Collapse
|
32
|
Jain MS, Polanski K, Conde CD, Chen X, Park J, Mamanova L, Knights A, Botting RA, Stephenson E, Haniffa M, Lamacraft A, Efremova M, Teichmann SA. MultiMAP: dimensionality reduction and integration of multimodal data. Genome Biol 2021; 22:346. [PMID: 34930412 PMCID: PMC8686224 DOI: 10.1186/s13059-021-02565-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/03/2021] [Indexed: 01/04/2023] Open
Abstract
Multimodal data is rapidly growing in many fields of science and engineering, including single-cell biology. We introduce MultiMAP, a novel algorithm for dimensionality reduction and integration. MultiMAP can integrate any number of datasets, leverages features not present in all datasets, is not restricted to a linear mapping, allows the user to specify the influence of each dataset, and is extremely scalable to large datasets. We apply MultiMAP to single-cell transcriptomics, chromatin accessibility, methylation, and spatial data and show that it outperforms current approaches. On a new thymus dataset, we use MultiMAP to integrate cells along a temporal trajectory. This enables quantitative comparison of transcription factor expression and binding site accessibility over the course of T cell differentiation, revealing patterns of expression versus binding site opening kinetics.
Collapse
Affiliation(s)
- Mika Sarkin Jain
- Theory of Condensed Matter, Dept Physics, Cavendish Laboratory, University of Cambridge, JJ Thomson Ave, Cambridge, CB3 0HE, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| | - Krzysztof Polanski
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | | | - Xi Chen
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Southern University of Science and Technology, 1088 Xueyuan Ave, Nanshan, Shenzhen, 518055, Guangdong Province, China
| | - Jongeun Park
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- KAIST, 291 Daehak-ro, Eoeun-dong, Yuseong-gu, Daejeon, South Korea
| | - Lira Mamanova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Andrew Knights
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Rachel A Botting
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Emily Stephenson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Austen Lamacraft
- Theory of Condensed Matter, Dept Physics, Cavendish Laboratory, University of Cambridge, JJ Thomson Ave, Cambridge, CB3 0HE, UK
| | - Mirjana Efremova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
- Barts Cancer Institute, Queen Mary University of London, London, UK.
| | - Sarah A Teichmann
- Theory of Condensed Matter, Dept Physics, Cavendish Laboratory, University of Cambridge, JJ Thomson Ave, Cambridge, CB3 0HE, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| |
Collapse
|
33
|
Nano PR, Nguyen CV, Mil J, Bhaduri A. Cortical Cartography: Mapping Arealization Using Single-Cell Omics Technology. Front Neural Circuits 2021; 15:788560. [PMID: 34955761 PMCID: PMC8707733 DOI: 10.3389/fncir.2021.788560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
The cerebral cortex derives its cognitive power from a modular network of specialized areas processing a multitude of information. The assembly and organization of these regions is vital for human behavior and perception, as evidenced by the prevalence of area-specific phenotypes that manifest in neurodevelopmental and psychiatric disorders. Generations of scientists have examined the architecture of the human cortex, but efforts to capture the gene networks which drive arealization have been hampered by the lack of tractable models of human neurodevelopment. Advancements in "omics" technologies, imaging, and computational power have enabled exciting breakthroughs into the molecular and structural characteristics of cortical areas, including transcriptomic, epigenomic, metabolomic, and proteomic profiles of mammalian models. Here we review the single-omics atlases that have shaped our current understanding of cortical areas, and their potential to fuel a new era of multi-omic single-cell endeavors to interrogate both the developing and adult human cortex.
Collapse
Affiliation(s)
| | | | | | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
34
|
Bakken TE, Jorstad NL, Hu Q, Lake BB, Tian W, Kalmbach BE, Crow M, Hodge RD, Krienen FM, Sorensen SA, Eggermont J, Yao Z, Aevermann BD, Aldridge AI, Bartlett A, Bertagnolli D, Casper T, Castanon RG, Crichton K, Daigle TL, Dalley R, Dee N, Dembrow N, Diep D, Ding SL, Dong W, Fang R, Fischer S, Goldman M, Goldy J, Graybuck LT, Herb BR, Hou X, Kancherla J, Kroll M, Lathia K, van Lew B, Li YE, Liu CS, Liu H, Lucero JD, Mahurkar A, McMillen D, Miller JA, Moussa M, Nery JR, Nicovich PR, Niu SY, Orvis J, Osteen JK, Owen S, Palmer CR, Pham T, Plongthongkum N, Poirion O, Reed NM, Rimorin C, Rivkin A, Romanow WJ, Sedeño-Cortés AE, Siletti K, Somasundaram S, Sulc J, Tieu M, Torkelson A, Tung H, Wang X, Xie F, Yanny AM, Zhang R, Ament SA, Behrens MM, Bravo HC, Chun J, Dobin A, Gillis J, Hertzano R, Hof PR, Höllt T, Horwitz GD, Keene CD, Kharchenko PV, Ko AL, Lelieveldt BP, Luo C, Mukamel EA, Pinto-Duarte A, Preissl S, Regev A, Ren B, Scheuermann RH, Smith K, Spain WJ, White OR, Koch C, Hawrylycz M, Tasic B, Macosko EZ, McCarroll SA, Ting JT, et alBakken TE, Jorstad NL, Hu Q, Lake BB, Tian W, Kalmbach BE, Crow M, Hodge RD, Krienen FM, Sorensen SA, Eggermont J, Yao Z, Aevermann BD, Aldridge AI, Bartlett A, Bertagnolli D, Casper T, Castanon RG, Crichton K, Daigle TL, Dalley R, Dee N, Dembrow N, Diep D, Ding SL, Dong W, Fang R, Fischer S, Goldman M, Goldy J, Graybuck LT, Herb BR, Hou X, Kancherla J, Kroll M, Lathia K, van Lew B, Li YE, Liu CS, Liu H, Lucero JD, Mahurkar A, McMillen D, Miller JA, Moussa M, Nery JR, Nicovich PR, Niu SY, Orvis J, Osteen JK, Owen S, Palmer CR, Pham T, Plongthongkum N, Poirion O, Reed NM, Rimorin C, Rivkin A, Romanow WJ, Sedeño-Cortés AE, Siletti K, Somasundaram S, Sulc J, Tieu M, Torkelson A, Tung H, Wang X, Xie F, Yanny AM, Zhang R, Ament SA, Behrens MM, Bravo HC, Chun J, Dobin A, Gillis J, Hertzano R, Hof PR, Höllt T, Horwitz GD, Keene CD, Kharchenko PV, Ko AL, Lelieveldt BP, Luo C, Mukamel EA, Pinto-Duarte A, Preissl S, Regev A, Ren B, Scheuermann RH, Smith K, Spain WJ, White OR, Koch C, Hawrylycz M, Tasic B, Macosko EZ, McCarroll SA, Ting JT, Zeng H, Zhang K, Feng G, Ecker JR, Linnarsson S, Lein ES. Comparative cellular analysis of motor cortex in human, marmoset and mouse. Nature 2021; 598:111-119. [PMID: 34616062 PMCID: PMC8494640 DOI: 10.1038/s41586-021-03465-8] [Show More Authors] [Citation(s) in RCA: 410] [Impact Index Per Article: 102.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 03/17/2021] [Indexed: 12/11/2022]
Abstract
The primary motor cortex (M1) is essential for voluntary fine-motor control and is functionally conserved across mammals1. Here, using high-throughput transcriptomic and epigenomic profiling of more than 450,000 single nuclei in humans, marmoset monkeys and mice, we demonstrate a broadly conserved cellular makeup of this region, with similarities that mirror evolutionary distance and are consistent between the transcriptome and epigenome. The core conserved molecular identities of neuronal and non-neuronal cell types allow us to generate a cross-species consensus classification of cell types, and to infer conserved properties of cell types across species. Despite the overall conservation, however, many species-dependent specializations are apparent, including differences in cell-type proportions, gene expression, DNA methylation and chromatin state. Few cell-type marker genes are conserved across species, revealing a short list of candidate genes and regulatory mechanisms that are responsible for conserved features of homologous cell types, such as the GABAergic chandelier cells. This consensus transcriptomic classification allows us to use patch-seq (a combination of whole-cell patch-clamp recordings, RNA sequencing and morphological characterization) to identify corticospinal Betz cells from layer 5 in non-human primates and humans, and to characterize their highly specialized physiology and anatomy. These findings highlight the robust molecular underpinnings of cell-type diversity in M1 across mammals, and point to the genes and regulatory pathways responsible for the functional identity of cell types and their species-specific adaptations.
Collapse
Affiliation(s)
| | | | - Qiwen Hu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Blue B Lake
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Wei Tian
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Brian E Kalmbach
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Megan Crow
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Fenna M Krienen
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | - Jeroen Eggermont
- LKEB, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Andrew I Aldridge
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Anna Bartlett
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | | | - Rosa G Castanon
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | | | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nikolai Dembrow
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
- Epilepsy Center of Excellence, Department of Veterans Affairs Medical Center, Seattle, WA, USA
| | - Dinh Diep
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | - Weixiu Dong
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Rongxin Fang
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Stephan Fischer
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Melissa Goldman
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Brian R Herb
- Institute for Genomes Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xiaomeng Hou
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jayaram Kancherla
- Department of Computer Science, University of Maryland College Park, College Park, MD, USA
| | | | - Kanan Lathia
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Baldur van Lew
- LKEB, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yang Eric Li
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Christine S Liu
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Biomedical Sciences Program, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Hanqing Liu
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Anup Mahurkar
- Institute for Genomes Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | - Joseph R Nery
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Sheng-Yong Niu
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Computer Science and Engineering Program, University of California, San Diego, La Jolla, CA, USA
| | - Joshua Orvis
- Institute for Genomes Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Julia K Osteen
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Scott Owen
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Carter R Palmer
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Biomedical Sciences Program, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Thanh Pham
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nongluk Plongthongkum
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Olivier Poirion
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Nora M Reed
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | - Angeline Rivkin
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - William J Romanow
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Kimberly Siletti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Xinxin Wang
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Fangming Xie
- Department of Physics, University of California, San Diego, La Jolla, CA, USA
| | | | - Renee Zhang
- J. Craig Venter Institute, La Jolla, CA, USA
| | - Seth A Ament
- Institute for Genomes Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Hector Corrada Bravo
- Department of Computer Science, University of Maryland College Park, College Park, MD, USA
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Jesse Gillis
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ronna Hertzano
- Departments of Otorhinolaryngology, Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas Höllt
- Computer Graphics and Visualization Group, Delt University of Technology, Delft, The Netherlands
| | - Gregory D Horwitz
- Department of Physiology and Biophysics, Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Peter V Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Andrew L Ko
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA
- Regional Epilepsy Center, Harborview Medical Center, Seattle, WA, USA
| | - Boudewijn P Lelieveldt
- LKEB, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Pattern Recognition and Bioinformatics group, Delft University of Technology, Delft, The Netherlands
| | - Chongyuan Luo
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Eran A Mukamel
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA
| | | | - Sebastian Preissl
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bing Ren
- Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Richard H Scheuermann
- J. Craig Venter Institute, La Jolla, CA, USA
- Department of Pathology, University of California, San Diego, CA, USA
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - William J Spain
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
- Epilepsy Center of Excellence, Department of Veterans Affairs Medical Center, Seattle, WA, USA
| | - Owen R White
- Institute for Genomes Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | - Steven A McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Kun Zhang
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joseph R Ecker
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sten Linnarsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA.
| |
Collapse
|
35
|
Peng H, Xie P, Liu L, Kuang X, Wang Y, Qu L, Gong H, Jiang S, Li A, Ruan Z, Ding L, Yao Z, Chen C, Chen M, Daigle TL, Dalley R, Ding Z, Duan Y, Feiner A, He P, Hill C, Hirokawa KE, Hong G, Huang L, Kebede S, Kuo HC, Larsen R, Lesnar P, Li L, Li Q, Li X, Li Y, Li Y, Liu A, Lu D, Mok S, Ng L, Nguyen TN, Ouyang Q, Pan J, Shen E, Song Y, Sunkin SM, Tasic B, Veldman MB, Wakeman W, Wan W, Wang P, Wang Q, Wang T, Wang Y, Xiong F, Xiong W, Xu W, Ye M, Yin L, Yu Y, Yuan J, Yuan J, Yun Z, Zeng S, Zhang S, Zhao S, Zhao Z, Zhou Z, Huang ZJ, Esposito L, Hawrylycz MJ, Sorensen SA, Yang XW, Zheng Y, Gu Z, Xie W, Koch C, Luo Q, Harris JA, Wang Y, Zeng H. Morphological diversity of single neurons in molecularly defined cell types. Nature 2021; 598:174-181. [PMID: 34616072 PMCID: PMC8494643 DOI: 10.1038/s41586-021-03941-1] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 08/24/2021] [Indexed: 12/23/2022]
Abstract
Dendritic and axonal morphology reflects the input and output of neurons and is a defining feature of neuronal types1,2, yet our knowledge of its diversity remains limited. Here, to systematically examine complete single-neuron morphologies on a brain-wide scale, we established a pipeline encompassing sparse labelling, whole-brain imaging, reconstruction, registration and analysis. We fully reconstructed 1,741 neurons from cortex, claustrum, thalamus, striatum and other brain regions in mice. We identified 11 major projection neuron types with distinct morphological features and corresponding transcriptomic identities. Extensive projectional diversity was found within each of these major types, on the basis of which some types were clustered into more refined subtypes. This diversity follows a set of generalizable principles that govern long-range axonal projections at different levels, including molecular correspondence, divergent or convergent projection, axon termination pattern, regional specificity, topography, and individual cell variability. Although clear concordance with transcriptomic profiles is evident at the level of major projection type, fine-grained morphological diversity often does not readily correlate with transcriptomic subtypes derived from unsupervised clustering, highlighting the need for single-cell cross-modality studies. Overall, our study demonstrates the crucial need for quantitative description of complete single-cell anatomy in cell-type classification, as single-cell morphological diversity reveals a plethora of ways in which different cell types and their individual members may contribute to the configuration and function of their respective circuits.
Collapse
Affiliation(s)
- Hanchuan Peng
- Allen Institute for Brain Science, Seattle, WA, USA.
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China.
| | - Peng Xie
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Lijuan Liu
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
- Ministry of Education Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Xiuli Kuang
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Yimin Wang
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Lei Qu
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
- Key Laboratory of Intelligent Computation and Signal Processing, Ministry of Education, Anhui University, Hefei, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, China
| | - Shengdian Jiang
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, China
| | - Zongcai Ruan
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Liya Ding
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Chao Chen
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Mengya Chen
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | | | | | - Zhangcan Ding
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Yanjun Duan
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Aaron Feiner
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Ping He
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Chris Hill
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Karla E Hirokawa
- Allen Institute for Brain Science, Seattle, WA, USA
- Cajal Neuroscience, Seattle, WA, USA
| | - Guodong Hong
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
- Ministry of Education Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Lei Huang
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Sara Kebede
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Phil Lesnar
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Longfei Li
- Key Laboratory of Intelligent Computation and Signal Processing, Ministry of Education, Anhui University, Hefei, China
| | - Qi Li
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Xiangning Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, China
| | - Yaoyao Li
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Yuanyuan Li
- Key Laboratory of Intelligent Computation and Signal Processing, Ministry of Education, Anhui University, Hefei, China
| | - An Liu
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
- Ministry of Education Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | | | | | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Thuc Nghi Nguyen
- Allen Institute for Brain Science, Seattle, WA, USA
- Cajal Neuroscience, Seattle, WA, USA
| | - Qiang Ouyang
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Jintao Pan
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Elise Shen
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Yuanyuan Song
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | | | | | - Matthew B Veldman
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Wan Wan
- Key Laboratory of Intelligent Computation and Signal Processing, Ministry of Education, Anhui University, Hefei, China
| | - Peng Wang
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Quanxin Wang
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tao Wang
- Key Laboratory of Intelligent Computation and Signal Processing, Ministry of Education, Anhui University, Hefei, China
| | - Yaping Wang
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Feng Xiong
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Wei Xiong
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Wenjie Xu
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Min Ye
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Lulu Yin
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Yang Yu
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jia Yuan
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
- Ministry of Education Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Jing Yuan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, China
| | - Zhixi Yun
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Shaoqun Zeng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
| | - Shichen Zhang
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Sujun Zhao
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Zijun Zhao
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Zhi Zhou
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Z Josh Huang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | | | | | | | - X William Yang
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Zhongze Gu
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Wei Xie
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, China
- Ministry of Education Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | | | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- School of Biomedical Engineering, Hainan University, Haikou, China
| | - Julie A Harris
- Allen Institute for Brain Science, Seattle, WA, USA
- Cajal Neuroscience, Seattle, WA, USA
| | - Yun Wang
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA.
| |
Collapse
|
36
|
Yao Z, Liu H, Xie F, Fischer S, Adkins RS, Aldridge AI, Ament SA, Bartlett A, Behrens MM, Van den Berge K, Bertagnolli D, de Bézieux HR, Biancalani T, Booeshaghi AS, Bravo HC, Casper T, Colantuoni C, Crabtree J, Creasy H, Crichton K, Crow M, Dee N, Dougherty EL, Doyle WI, Dudoit S, Fang R, Felix V, Fong O, Giglio M, Goldy J, Hawrylycz M, Herb BR, Hertzano R, Hou X, Hu Q, Kancherla J, Kroll M, Lathia K, Li YE, Lucero JD, Luo C, Mahurkar A, McMillen D, Nadaf NM, Nery JR, Nguyen TN, Niu SY, Ntranos V, Orvis J, Osteen JK, Pham T, Pinto-Duarte A, Poirion O, Preissl S, Purdom E, Rimorin C, Risso D, Rivkin AC, Smith K, Street K, Sulc J, Svensson V, Tieu M, Torkelson A, Tung H, Vaishnav ED, Vanderburg CR, van Velthoven C, Wang X, White OR, Huang ZJ, Kharchenko PV, Pachter L, Ngai J, Regev A, Tasic B, Welch JD, Gillis J, Macosko EZ, Ren B, Ecker JR, Zeng H, Mukamel EA. A transcriptomic and epigenomic cell atlas of the mouse primary motor cortex. Nature 2021; 598:103-110. [PMID: 34616066 PMCID: PMC8494649 DOI: 10.1038/s41586-021-03500-8] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 03/26/2021] [Indexed: 12/30/2022]
Abstract
Single-cell transcriptomics can provide quantitative molecular signatures for large, unbiased samples of the diverse cell types in the brain1-3. With the proliferation of multi-omics datasets, a major challenge is to validate and integrate results into a biological understanding of cell-type organization. Here we generated transcriptomes and epigenomes from more than 500,000 individual cells in the mouse primary motor cortex, a structure that has an evolutionarily conserved role in locomotion. We developed computational and statistical methods to integrate multimodal data and quantitatively validate cell-type reproducibility. The resulting reference atlas-containing over 56 neuronal cell types that are highly replicable across analysis methods, sequencing technologies and modalities-is a comprehensive molecular and genomic account of the diverse neuronal and non-neuronal cell types in the mouse primary motor cortex. The atlas includes a population of excitatory neurons that resemble pyramidal cells in layer 4 in other cortical regions4. We further discovered thousands of concordant marker genes and gene regulatory elements for these cell types. Our results highlight the complex molecular regulation of cell types in the brain and will directly enable the design of reagents to target specific cell types in the mouse primary motor cortex for functional analysis.
Collapse
Affiliation(s)
- Zizhen Yao
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Hanqing Liu
- grid.250671.70000 0001 0662 7144Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Fangming Xie
- grid.266100.30000 0001 2107 4242Department of Physics, University of California, San Diego, La Jolla, CA USA
| | - Stephan Fischer
- grid.225279.90000 0004 0387 3667Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY USA
| | - Ricky S. Adkins
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - Andrew I. Aldridge
- grid.250671.70000 0001 0662 7144Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Seth A. Ament
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - Anna Bartlett
- grid.250671.70000 0001 0662 7144Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - M. Margarita Behrens
- grid.250671.70000 0001 0662 7144Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Koen Van den Berge
- grid.47840.3f0000 0001 2181 7878Department of Statistics, University of California, Berkeley, Berkeley, CA USA ,grid.5342.00000 0001 2069 7798Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Gent, Belgium
| | | | - Hector Roux de Bézieux
- grid.47840.3f0000 0001 2181 7878Division of Biostatistics, School of Public Health, University of California, Berkeley, Berkeley, CA USA
| | | | - A. Sina Booeshaghi
- grid.20861.3d0000000107068890California Institute of Technology, Pasadena, CA USA
| | - Héctor Corrada Bravo
- grid.164295.d0000 0001 0941 7177Center for Bioinformatics and Computational Biology, University of Maryland, College Park, College Park, MD USA
| | - Tamara Casper
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Carlo Colantuoni
- grid.21107.350000 0001 2171 9311Johns Hopkins School of Medicine, Department of Neurology, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Johns Hopkins School of Medicine, Department of Neuroscience, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD USA
| | - Jonathan Crabtree
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - Heather Creasy
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | | | - Megan Crow
- grid.225279.90000 0004 0387 3667Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY USA
| | - Nick Dee
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | | | - Wayne I. Doyle
- grid.266100.30000 0001 2107 4242Department of Cognitive Science, University of California, San Diego, La Jolla, CA USA
| | - Sandrine Dudoit
- grid.47840.3f0000 0001 2181 7878Department of Statistics, University of California, Berkeley, Berkeley, CA USA
| | - Rongxin Fang
- grid.266100.30000 0001 2107 4242Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, San Diego, CA USA
| | - Victor Felix
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - Olivia Fong
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Michelle Giglio
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - Jeff Goldy
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Mike Hawrylycz
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Brian R. Herb
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - Ronna Hertzano
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Department of Otorhinolaryngology, Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Xiaomeng Hou
- grid.266100.30000 0001 2107 4242Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA USA
| | - Qiwen Hu
- grid.38142.3c000000041936754XDepartment of Biomedical Informatics, Harvard Medical School, Boston, MA USA
| | - Jayaram Kancherla
- grid.164295.d0000 0001 0941 7177Center for Bioinformatics and Computational Biology, University of Maryland, College Park, College Park, MD USA
| | - Matthew Kroll
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Kanan Lathia
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Yang Eric Li
- grid.1052.60000000097371625Ludwig Institute for Cancer Research, La Jolla, CA USA
| | - Jacinta D. Lucero
- grid.250671.70000 0001 0662 7144Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Chongyuan Luo
- grid.250671.70000 0001 0662 7144Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA USA ,grid.250671.70000 0001 0662 7144Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Anup Mahurkar
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | | | - Naeem M. Nadaf
- grid.66859.34Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Joseph R. Nery
- grid.250671.70000 0001 0662 7144Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA
| | | | - Sheng-Yong Niu
- grid.250671.70000 0001 0662 7144Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Vasilis Ntranos
- grid.266102.10000 0001 2297 6811University of California, San Francisco, San Francisco, CA USA
| | - Joshua Orvis
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - Julia K. Osteen
- grid.250671.70000 0001 0662 7144Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Thanh Pham
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Antonio Pinto-Duarte
- grid.250671.70000 0001 0662 7144Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Olivier Poirion
- grid.266100.30000 0001 2107 4242Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA USA
| | - Sebastian Preissl
- grid.266100.30000 0001 2107 4242Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA USA
| | - Elizabeth Purdom
- grid.47840.3f0000 0001 2181 7878Department of Statistics, University of California, Berkeley, Berkeley, CA USA
| | | | - Davide Risso
- grid.5608.b0000 0004 1757 3470Department of Statistical Sciences, University of Padova, Padova, Italy
| | - Angeline C. Rivkin
- grid.250671.70000 0001 0662 7144Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Kimberly Smith
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Kelly Street
- grid.65499.370000 0001 2106 9910Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA USA
| | - Josef Sulc
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Valentine Svensson
- grid.20861.3d0000000107068890California Institute of Technology, Pasadena, CA USA
| | - Michael Tieu
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Amy Torkelson
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Herman Tung
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | | | | | | | - Xinxin Wang
- grid.266100.30000 0001 2107 4242Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA USA ,grid.4367.60000 0001 2355 7002Present Address: McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO USA
| | - Owen R. White
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD USA
| | - Z. Josh Huang
- grid.225279.90000 0004 0387 3667Cold Spring Harbor Laboratory, Cold Spring Harbor, NY USA
| | - Peter V. Kharchenko
- grid.38142.3c000000041936754XDepartment of Biomedical Informatics, Harvard Medical School, Boston, MA USA
| | - Lior Pachter
- grid.20861.3d0000000107068890California Institute of Technology, Pasadena, CA USA
| | - John Ngai
- grid.47840.3f0000 0001 2181 7878Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Aviv Regev
- grid.66859.34Broad Institute of MIT and Harvard, Cambridge, MA USA ,grid.116068.80000 0001 2341 2786Howard Hughes Medical Institute, Department of Biology, MIT, Cambridge, MA USA
| | - Bosiljka Tasic
- grid.417881.3Allen Institute for Brain Science, Seattle, WA USA
| | - Joshua D. Welch
- grid.214458.e0000000086837370Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI USA
| | - Jesse Gillis
- grid.225279.90000 0004 0387 3667Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY USA
| | - Evan Z. Macosko
- grid.66859.34Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Bing Ren
- grid.266100.30000 0001 2107 4242Center for Epigenomics, Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA USA ,grid.1052.60000000097371625Ludwig Institute for Cancer Research, La Jolla, CA USA
| | - Joseph R. Ecker
- grid.250671.70000 0001 0662 7144Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA USA ,grid.250671.70000 0001 0662 7144Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA.
| | - Eran A. Mukamel
- grid.266100.30000 0001 2107 4242Department of Cognitive Science, University of California, San Diego, La Jolla, CA USA
| |
Collapse
|
37
|
Scala F, Kobak D, Bernabucci M, Bernaerts Y, Cadwell CR, Castro JR, Hartmanis L, Jiang X, Laturnus S, Miranda E, Mulherkar S, Tan ZH, Yao Z, Zeng H, Sandberg R, Berens P, Tolias AS. Phenotypic variation of transcriptomic cell types in mouse motor cortex. Nature 2021; 598:144-150. [PMID: 33184512 PMCID: PMC8113357 DOI: 10.1038/s41586-020-2907-3] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022]
Abstract
Cortical neurons exhibit extreme diversity in gene expression as well as in morphological and electrophysiological properties1,2. Most existing neural taxonomies are based on either transcriptomic3,4 or morpho-electric5,6 criteria, as it has been technically challenging to study both aspects of neuronal diversity in the same set of cells7. Here we used Patch-seq8 to combine patch-clamp recording, biocytin staining, and single-cell RNA sequencing of more than 1,300 neurons in adult mouse primary motor cortex, providing a morpho-electric annotation of almost all transcriptomically defined neural cell types. We found that, although broad families of transcriptomic types (those expressing Vip, Pvalb, Sst and so on) had distinct and essentially non-overlapping morpho-electric phenotypes, individual transcriptomic types within the same family were not well separated in the morpho-electric space. Instead, there was a continuum of variability in morphology and electrophysiology, with neighbouring transcriptomic cell types showing similar morpho-electric features, often without clear boundaries between them. Our results suggest that neuronal types in the neocortex do not always form discrete entities. Instead, neurons form a hierarchy that consists of distinct non-overlapping branches at the level of families, but can form continuous and correlated transcriptomic and morpho-electrical landscapes within families.
Collapse
Affiliation(s)
- Federico Scala
- Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Dmitry Kobak
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Matteo Bernabucci
- Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Yves Bernaerts
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- International Max Planck Research School for Intelligent Systems, Tübingen, Germany
| | - Cathryn René Cadwell
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Jesus Ramon Castro
- Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Leonard Hartmanis
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Xiaolong Jiang
- Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Houston, TX, USA
| | - Sophie Laturnus
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Elanine Miranda
- Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Zheng Huan Tan
- Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Rickard Sandberg
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Philipp Berens
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany.
- Center for Integrative Neuroscience, University of Tübingen, Tübingen, Germany.
- Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany.
- Bernstein Center for Computational Neuroscience, University of Tübingen, Tübingen, Germany.
| | - Andreas S Tolias
- Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
38
|
Zhang M, Eichhorn SW, Zingg B, Yao Z, Cotter K, Zeng H, Dong H, Zhuang X. Spatially resolved cell atlas of the mouse primary motor cortex by MERFISH. Nature 2021; 598:137-143. [PMID: 34616063 PMCID: PMC8494645 DOI: 10.1038/s41586-021-03705-x] [Citation(s) in RCA: 260] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/08/2021] [Indexed: 11/09/2022]
Abstract
A mammalian brain is composed of numerous cell types organized in an intricate manner to form functional neural circuits. Single-cell RNA sequencing allows systematic identification of cell types based on their gene expression profiles and has revealed many distinct cell populations in the brain1,2. Single-cell epigenomic profiling3,4 further provides information on gene-regulatory signatures of different cell types. Understanding how different cell types contribute to brain function, however, requires knowledge of their spatial organization and connectivity, which is not preserved in sequencing-based methods that involve cell dissociation. Here we used a single-cell transcriptome-imaging method, multiplexed error-robust fluorescence in situ hybridization (MERFISH)5, to generate a molecularly defined and spatially resolved cell atlas of the mouse primary motor cortex. We profiled approximately 300,000 cells in the mouse primary motor cortex and its adjacent areas, identified 95 neuronal and non-neuronal cell clusters, and revealed a complex spatial map in which not only excitatory but also most inhibitory neuronal clusters adopted laminar organizations. Intratelencephalic neurons formed a largely continuous gradient along the cortical depth axis, in which the gene expression of individual cells correlated with their cortical depths. Furthermore, we integrated MERFISH with retrograde labelling to probe projection targets of neurons of the mouse primary motor cortex and found that their cortical projections formed a complex network in which individual neuronal clusters project to multiple target regions and individual target regions receive inputs from multiple neuronal clusters.
Collapse
Affiliation(s)
- Meng Zhang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Physics, Harvard University, Cambridge, MA, USA
| | - Stephen W Eichhorn
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Physics, Harvard University, Cambridge, MA, USA
| | - Brian Zingg
- Center for Integrative Connectomics, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- UCLA Brain Research & Artificial Intelligence Nexus, Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Kaelan Cotter
- Center for Integrative Connectomics, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- UCLA Brain Research & Artificial Intelligence Nexus, Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongwei Dong
- Center for Integrative Connectomics, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- UCLA Brain Research & Artificial Intelligence Nexus, Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Department of Physics, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
39
|
A multimodal cell census and atlas of the mammalian primary motor cortex. Nature 2021; 598:86-102. [PMID: 34616075 DOI: 10.1101/2020.10.19.343129v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 08/25/2021] [Indexed: 05/26/2023]
Abstract
Here we report the generation of a multimodal cell census and atlas of the mammalian primary motor cortex as the initial product of the BRAIN Initiative Cell Census Network (BICCN). This was achieved by coordinated large-scale analyses of single-cell transcriptomes, chromatin accessibility, DNA methylomes, spatially resolved single-cell transcriptomes, morphological and electrophysiological properties and cellular resolution input-output mapping, integrated through cross-modal computational analysis. Our results advance the collective knowledge and understanding of brain cell-type organization1-5. First, our study reveals a unified molecular genetic landscape of cortical cell types that integrates their transcriptome, open chromatin and DNA methylation maps. Second, cross-species analysis achieves a consensus taxonomy of transcriptomic types and their hierarchical organization that is conserved from mouse to marmoset and human. Third, in situ single-cell transcriptomics provides a spatially resolved cell-type atlas of the motor cortex. Fourth, cross-modal analysis provides compelling evidence for the transcriptomic, epigenomic and gene regulatory basis of neuronal phenotypes such as their physiological and anatomical properties, demonstrating the biological validity and genomic underpinning of neuron types. We further present an extensive genetic toolset for targeting glutamatergic neuron types towards linking their molecular and developmental identity to their circuit function. Together, our results establish a unifying and mechanistic framework of neuronal cell-type organization that integrates multi-layered molecular genetic and spatial information with multi-faceted phenotypic properties.
Collapse
|
40
|
BRAIN Initiative Cell Census Network (BICCN), Callaway EM, Dong HW, Ecker JR, Hawrylycz MJ, Huang ZJ, Lein ES, Ngai J, Osten P, Ren B, Tolias AS, White O, Zeng H, Zhuang X, Ascoli GA, Behrens MM, Chun J, Feng G, Gee JC, Ghosh SS, Halchenko YO, Hertzano R, Lim BK, Martone ME, Ng L, Pachter L, Ropelewski AJ, Tickle TL, Yang XW, Zhang K, Bakken TE, Berens P, Daigle TL, Harris JA, Jorstad NL, Kalmbach BE, Kobak D, Li YE, Liu H, Matho KS, Mukamel EA, Naeemi M, Scala F, Tan P, Ting JT, Xie F, Zhang M, Zhang Z, Zhou J, Zingg B, Armand E, Yao Z, Bertagnolli D, Casper T, Crichton K, Dee N, Diep D, Ding SL, Dong W, Dougherty EL, Fong O, Goldman M, Goldy J, Hodge RD, Hu L, Keene CD, Krienen FM, Kroll M, Lake BB, Lathia K, Linnarsson S, Liu CS, Macosko EZ, McCarroll SA, McMillen D, Nadaf NM, Nguyen TN, Palmer CR, Pham T, Plongthongkum N, Reed NM, Regev A, Rimorin C, Romanow WJ, Savoia S, Siletti K, Smith K, Sulc J, Tasic B, Tieu M, Torkelson A, Tung H, van Velthoven CTJ, Vanderburg CR, Yanny AM, Fang R, Hou X, Lucero JD, Osteen JK, Pinto-Duarte A, et alBRAIN Initiative Cell Census Network (BICCN), Callaway EM, Dong HW, Ecker JR, Hawrylycz MJ, Huang ZJ, Lein ES, Ngai J, Osten P, Ren B, Tolias AS, White O, Zeng H, Zhuang X, Ascoli GA, Behrens MM, Chun J, Feng G, Gee JC, Ghosh SS, Halchenko YO, Hertzano R, Lim BK, Martone ME, Ng L, Pachter L, Ropelewski AJ, Tickle TL, Yang XW, Zhang K, Bakken TE, Berens P, Daigle TL, Harris JA, Jorstad NL, Kalmbach BE, Kobak D, Li YE, Liu H, Matho KS, Mukamel EA, Naeemi M, Scala F, Tan P, Ting JT, Xie F, Zhang M, Zhang Z, Zhou J, Zingg B, Armand E, Yao Z, Bertagnolli D, Casper T, Crichton K, Dee N, Diep D, Ding SL, Dong W, Dougherty EL, Fong O, Goldman M, Goldy J, Hodge RD, Hu L, Keene CD, Krienen FM, Kroll M, Lake BB, Lathia K, Linnarsson S, Liu CS, Macosko EZ, McCarroll SA, McMillen D, Nadaf NM, Nguyen TN, Palmer CR, Pham T, Plongthongkum N, Reed NM, Regev A, Rimorin C, Romanow WJ, Savoia S, Siletti K, Smith K, Sulc J, Tasic B, Tieu M, Torkelson A, Tung H, van Velthoven CTJ, Vanderburg CR, Yanny AM, Fang R, Hou X, Lucero JD, Osteen JK, Pinto-Duarte A, Poirion O, Preissl S, Wang X, Aldridge AI, Bartlett A, Boggeman L, O’Connor C, Castanon RG, Chen H, Fitzpatrick C, Luo C, Nery JR, Nunn M, Rivkin AC, Tian W, Dominguez B, Ito-Cole T, Jacobs M, Jin X, Lee CT, Lee KF, Miyazaki PA, Pang Y, Rashid M, Smith JB, Vu M, Williams E, Biancalani T, Booeshaghi AS, Crow M, Dudoit S, Fischer S, Gillis J, Hu Q, Kharchenko PV, Niu SY, Ntranos V, Purdom E, Risso D, de Bézieux HR, Somasundaram S, Street K, Svensson V, Vaishnav ED, Van den Berge K, Welch JD, An X, Bateup HS, Bowman I, Chance RK, Foster NN, Galbavy W, Gong H, Gou L, Hatfield JT, Hintiryan H, Hirokawa KE, Kim G, Kramer DJ, Li A, Li X, Luo Q, Muñoz-Castañeda R, Stafford DA, Feng Z, Jia X, Jiang S, Jiang T, Kuang X, Larsen R, Lesnar P, Li Y, Li Y, Liu L, Peng H, Qu L, Ren M, Ruan Z, Shen E, Song Y, Wakeman W, Wang P, Wang Y, Wang Y, Yin L, Yuan J, Zhao S, Zhao X, Narasimhan A, Palaniswamy R, Banerjee S, Ding L, Huilgol D, Huo B, Kuo HC, Laturnus S, Li X, Mitra PP, Mizrachi J, Wang Q, Xie P, Xiong F, Yu Y, Eichhorn SW, Berg J, Bernabucci M, Bernaerts Y, Cadwell CR, Castro JR, Dalley R, Hartmanis L, Horwitz GD, Jiang X, Ko AL, Miranda E, Mulherkar S, Nicovich PR, Owen SF, Sandberg R, Sorensen SA, Tan ZH, Allen S, Hockemeyer D, Lee AY, Veldman MB, Adkins RS, Ament SA, Bravo HC, Carter R, Chatterjee A, Colantuoni C, Crabtree J, Creasy H, Felix V, Giglio M, Herb BR, Kancherla J, Mahurkar A, McCracken C, Nickel L, Olley D, Orvis J, Schor M, Hood G, Dichter B, Grauer M, Helba B, Bandrowski A, Barkas N, Carlin B, D’Orazi FD, Degatano K, Gillespie TH, Khajouei F, Konwar K, Thompson C, Kelly K, Mok S, Sunkin S, BRAIN Initiative Cell Census Network (BICCN) Corresponding authors, BICCN contributing principal investigators, Principal manuscript editors, Manuscript writing and figure generation, Analysis coordination, Integrated data analysis, scRNA-seq and snRNA-seq data generation and processing, ATAC-seq data generation and processing, Methylcytosine data production and analysis, Epi-retro-seq data generation and processing, ‘Omics data analysis, Tracing and connectivity data generation, Morphology data generation and reconstruction, OLST/STPT and other data generation, Morphology, connectivity and imaging analysis, Spatially resolved single-cell transcriptomics (MERFISH), Multimodal profiling (Patch-seq), Transgenic tools, NeMO archive and analytics, Brain Image Library (BIL) archive, DANDI archive, Brain Cell Data Center (BCDC), Project management. A multimodal cell census and atlas of the mammalian primary motor cortex. Nature 2021; 598:86-102. [PMID: 34616075 PMCID: PMC8494634 DOI: 10.1038/s41586-021-03950-0] [Show More Authors] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 08/25/2021] [Indexed: 12/14/2022]
Abstract
Here we report the generation of a multimodal cell census and atlas of the mammalian primary motor cortex as the initial product of the BRAIN Initiative Cell Census Network (BICCN). This was achieved by coordinated large-scale analyses of single-cell transcriptomes, chromatin accessibility, DNA methylomes, spatially resolved single-cell transcriptomes, morphological and electrophysiological properties and cellular resolution input-output mapping, integrated through cross-modal computational analysis. Our results advance the collective knowledge and understanding of brain cell-type organization1-5. First, our study reveals a unified molecular genetic landscape of cortical cell types that integrates their transcriptome, open chromatin and DNA methylation maps. Second, cross-species analysis achieves a consensus taxonomy of transcriptomic types and their hierarchical organization that is conserved from mouse to marmoset and human. Third, in situ single-cell transcriptomics provides a spatially resolved cell-type atlas of the motor cortex. Fourth, cross-modal analysis provides compelling evidence for the transcriptomic, epigenomic and gene regulatory basis of neuronal phenotypes such as their physiological and anatomical properties, demonstrating the biological validity and genomic underpinning of neuron types. We further present an extensive genetic toolset for targeting glutamatergic neuron types towards linking their molecular and developmental identity to their circuit function. Together, our results establish a unifying and mechanistic framework of neuronal cell-type organization that integrates multi-layered molecular genetic and spatial information with multi-faceted phenotypic properties.
Collapse
|
41
|
Brennan EKW, Jedrasiak-Cape I, Kailasa S, Rice SP, Sudhakar SK, Ahmed OJ. Thalamus and claustrum control parallel layer 1 circuits in retrosplenial cortex. eLife 2021; 10:e62207. [PMID: 34170817 PMCID: PMC8233040 DOI: 10.7554/elife.62207] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 05/17/2021] [Indexed: 11/13/2022] Open
Abstract
The granular retrosplenial cortex (RSG) is critical for both spatial and non-spatial behaviors, but the underlying neural codes remain poorly understood. Here, we use optogenetic circuit mapping in mice to reveal a double dissociation that allows parallel circuits in superficial RSG to process disparate inputs. The anterior thalamus and dorsal subiculum, sources of spatial information, strongly and selectively recruit small low-rheobase (LR) pyramidal cells in RSG. In contrast, neighboring regular-spiking (RS) cells are preferentially controlled by claustral and anterior cingulate inputs, sources of mostly non-spatial information. Precise sublaminar axonal and dendritic arborization within RSG layer 1, in particular, permits this parallel processing. Observed thalamocortical synaptic dynamics enable computational models of LR neurons to compute the speed of head rotation, despite receiving head direction inputs that do not explicitly encode speed. Thus, parallel input streams identify a distinct principal neuronal subtype ideally positioned to support spatial orientation computations in the RSG.
Collapse
Affiliation(s)
- Ellen KW Brennan
- Department of Psychology, University of MichiganAnn ArborUnited States
- Neuroscience Graduate Program, University of MichiganAnn ArborUnited States
| | | | - Sameer Kailasa
- Department of Mathematics, University of MichiganAnn ArborUnited States
| | - Sharena P Rice
- Department of Psychology, University of MichiganAnn ArborUnited States
- Neuroscience Graduate Program, University of MichiganAnn ArborUnited States
| | | | - Omar J Ahmed
- Department of Psychology, University of MichiganAnn ArborUnited States
- Neuroscience Graduate Program, University of MichiganAnn ArborUnited States
- Michigan Center for Integrative Research in Critical Care, University of MichiganAnn ArborUnited States
- Kresge Hearing Research Institute, University of MichiganAnn ArborUnited States
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
| |
Collapse
|
42
|
Villalba RM, Behnke JA, Pare JF, Smith Y. Comparative Ultrastructural Analysis of Thalamocortical Innervation of the Primary Motor Cortex and Supplementary Motor Area in Control and MPTP-Treated Parkinsonian Monkeys. Cereb Cortex 2021; 31:3408-3425. [PMID: 33676368 PMCID: PMC8599722 DOI: 10.1093/cercor/bhab020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/29/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022] Open
Abstract
The synaptic organization of thalamic inputs to motor cortices remains poorly understood in primates. Thus, we compared the regional and synaptic connections of vGluT2-positive thalamocortical glutamatergic terminals in the supplementary motor area (SMA) and the primary motor cortex (M1) between control and MPTP-treated parkinsonian monkeys. In controls, vGluT2-containing fibers and terminal-like profiles invaded layer II-III and Vb of M1 and SMA. A significant reduction of vGluT2 labeling was found in layer Vb, but not in layer II-III, of parkinsonian animals, suggesting a potential thalamic denervation of deep cortical layers in parkinsonism. There was a significant difference in the pattern of synaptic connectivity in layers II-III, but not in layer Vb, between M1 and SMA of control monkeys. However, this difference was abolished in parkinsonian animals. No major difference was found in the proportion of perforated versus macular post-synaptic densities at thalamocortical synapses between control and parkinsonian monkeys in both cortical regions, except for a slight increase in the prevalence of perforated axo-dendritic synapses in the SMA of parkinsonian monkeys. Our findings suggest that disruption of the thalamic innervation of M1 and SMA may underlie pathophysiological changes of the motor thalamocortical loop in the state of parkinsonism.
Collapse
Affiliation(s)
- Rosa M Villalba
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
| | - Joseph A Behnke
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
| | - Jean-Francois Pare
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
| | - Yoland Smith
- Division of Neuropharmacology and Neurological Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- UDALL Center for Excellence for Parkinson’s Disease, Emory University, Atlanta, GA 30329, USA
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA 30329, USA
| |
Collapse
|
43
|
Anastasiades PG, Carter AG. Circuit organization of the rodent medial prefrontal cortex. Trends Neurosci 2021; 44:550-563. [PMID: 33972100 DOI: 10.1016/j.tins.2021.03.006] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 03/12/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022]
Abstract
The prefrontal cortex (PFC) orchestrates higher brain function and becomes disrupted in many mental health disorders. The rodent medial PFC (mPFC) possesses an enormous variety of projection neurons and interneurons. These cells are engaged by long-range inputs from other brain regions involved in cognition, motivation, and emotion. They also communicate in the local network via specific connections between excitatory and inhibitory cells. In this review, we describe the cellular diversity of the rodent mPFC, the impact of long-range afferents, and the specificity of local microcircuits. We highlight similarities with and differences between other cortical areas, illustrating how the circuit organization of the mPFC may give rise to its unique functional roles.
Collapse
Affiliation(s)
- Paul G Anastasiades
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Adam G Carter
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA.
| |
Collapse
|
44
|
Untangling the cortico-thalamo-cortical loop: cellular pieces of a knotty circuit puzzle. Nat Rev Neurosci 2021; 22:389-406. [PMID: 33958775 DOI: 10.1038/s41583-021-00459-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 12/22/2022]
Abstract
Functions of the neocortex depend on its bidirectional communication with the thalamus, via cortico-thalamo-cortical (CTC) loops. Recent work dissecting the synaptic connectivity in these loops is generating a clearer picture of their cellular organization. Here, we review findings across sensory, motor and cognitive areas, focusing on patterns of cell type-specific synaptic connections between the major types of cortical and thalamic neurons. We outline simple and complex CTC loops, and note features of these loops that appear to be general versus specialized. CTC loops are tightly interlinked with local cortical and corticocortical (CC) circuits, forming extended chains of loops that are probably critical for communication across hierarchically organized cerebral networks. Such CTC-CC loop chains appear to constitute a modular unit of organization, serving as scaffolding for area-specific structural and functional modifications. Inhibitory neurons and circuits are embedded throughout CTC loops, shaping the flow of excitation. We consider recent findings in the context of established CTC and CC circuit models, and highlight current efforts to pinpoint cell type-specific mechanisms in CTC loops involved in consciousness and perception. As pieces of the connectivity puzzle fall increasingly into place, this knowledge can guide further efforts to understand structure-function relationships in CTC loops.
Collapse
|
45
|
Vasistha NA, Khodosevich K. The impact of (ab)normal maternal environment on cortical development. Prog Neurobiol 2021; 202:102054. [PMID: 33905709 DOI: 10.1016/j.pneurobio.2021.102054] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/01/2021] [Accepted: 04/20/2021] [Indexed: 12/24/2022]
Abstract
The cortex in the mammalian brain is the most complex brain region that integrates sensory information and coordinates motor and cognitive processes. To perform such functions, the cortex contains multiple subtypes of neurons that are generated during embryogenesis. Newly born neurons migrate to their proper location in the cortex, grow axons and dendrites, and form neuronal circuits. These developmental processes in the fetal brain are regulated to a large extent by a great variety of factors derived from the mother - starting from simple nutrients as building blocks and ending with hormones. Thus, when the normal maternal environment is disturbed due to maternal infection, stress, malnutrition, or toxic substances, it might have a profound impact on cortical development and the offspring can develop a variety of neurodevelopmental disorders. Here we first describe the major developmental processes which generate neuronal diversity in the cortex. We then review our knowledge of how most common maternal insults affect cortical development, perturb neuronal circuits, and lead to neurodevelopmental disorders. We further present a concept of selective vulnerability of cortical neuronal subtypes to maternal-derived insults, where the vulnerability of cortical neurons and their progenitors to an insult depends on the time (developmental period), place (location in the developing brain), and type (unique features of a cell type and an insult). Finally, we provide evidence for the existence of selective vulnerability during cortical development and identify the most vulnerable neuronal types, stages of differentiation, and developmental time for major maternal-derived insults.
Collapse
Affiliation(s)
- Navneet A Vasistha
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
46
|
Yamawaki N, Raineri Tapies MG, Stults A, Smith GA, Shepherd GMG. Circuit organization of the excitatory sensorimotor loop through hand/forelimb S1 and M1. eLife 2021; 10:e66836. [PMID: 33851917 PMCID: PMC8046433 DOI: 10.7554/elife.66836] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/03/2021] [Indexed: 12/16/2022] Open
Abstract
Sensory-guided limb control relies on communication across sensorimotor loops. For active touch with the hand, the longest loop is the transcortical continuation of ascending pathways, particularly the lemnisco-cortical and corticocortical pathways carrying tactile signals via the cuneate nucleus, ventral posterior lateral (VPL) thalamus, and primary somatosensory (S1) and motor (M1) cortices to reach corticospinal neurons and influence descending activity. We characterized excitatory connectivity along this pathway in the mouse. In the lemnisco-cortical leg, disynaptic cuneate→VPL→S1 connections excited mainly layer (L) 4 neurons. In the corticocortical leg, S1→M1 connections from L2/3 and L5A neurons mainly excited downstream L2/3 neurons, which excite corticospinal neurons. The findings provide a detailed new wiring diagram for the hand/forelimb-related transcortical circuit, delineating a basic but complex set of cell-type-specific feedforward excitatory connections that selectively and extensively engage diverse intratelencephalic projection neurons, thereby polysynaptically linking subcortical somatosensory input to cortical motor output to spinal cord.
Collapse
Affiliation(s)
- Naoki Yamawaki
- Department of Physiology, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | | | - Austin Stults
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Gregory A Smith
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Gordon MG Shepherd
- Department of Physiology, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| |
Collapse
|
47
|
Farokhniaee A, Lowery MM. Cortical network effects of subthalamic deep brain stimulation in a thalamo-cortical microcircuit model. J Neural Eng 2021; 18. [DOI: 10.1088/1741-2552/abee50] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/12/2021] [Indexed: 11/12/2022]
|
48
|
Multiscale modeling of cortical gradients: The role of mesoscale circuits for linking macro- and microscale gradients of cortical organization and hierarchical information processing. Neuroimage 2021; 232:117846. [PMID: 33636345 DOI: 10.1016/j.neuroimage.2021.117846] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 12/16/2020] [Accepted: 02/04/2021] [Indexed: 11/21/2022] Open
Abstract
The gradient concept in neuroscience describes systematic and continuous progressions of features of cortical organization across the entire cortex. Recent multimodal studies revealed a macroscale gradient from primary sensory to transmodal association areas which is linked to increasing representational abstraction along the cortical hierarchy, and which is paralleled by microscale gradients of cytoarchitecture and gene expression profiles. Convergent or divergent evidence from these multimodal studies is then used to support inferences about the existence of one common or multiple scale-specific gradients of hierarchical information processing. This paper evaluates the validity of such inferences within the framework of multiscale modeling. In branches of physics and biology where multiscale modeling techniques are used, the simple averaging of microscale details can introduce errors in macroscale modeling if it ignores structures at the intermediate mesoscales of organization which affect system behavior. Conversely, information about mesoscale structures can be used to determine which microscale details are actually relevant to macroscale behavior. In this paper, I similarly argue that multiscale modeling of cortical gradients needs to take organization of mesoscale circuits into account if it affects the structure-function relation that the models describe. Information about these circuits provides crucial evidence for evaluating inferences from micro- and macroscale data to the role of cortical gradients in hierarchical information processing. My application of the multiscale modeling framework reveals that the gradient concept tracks multiple overlapping progressions of cortical properties, rather than one overall gradient of hierarchical information processing. I support this argument by proposing a mesoscale gradient of connectivity which describes architectural differences between granular and agranular circuits, and which helps us better understand the relation between neural connectivity and hierarchical information processing.
Collapse
|
49
|
Tang Q, Tsytsarev V, Yan F, Wang C, Erzurumlu RS, Chen Y. In vivo voltage-sensitive dye imaging of mouse cortical activity with mesoscopic optical tomography. NEUROPHOTONICS 2020; 7:041402. [PMID: 33274250 PMCID: PMC7708784 DOI: 10.1117/1.nph.7.4.041402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 11/11/2020] [Indexed: 05/11/2023]
Abstract
Significance: Cellular layering is a hallmark of the mammalian neocortex with layer and cell type-specific connections within the cortical mantle and subcortical connections. A key challenge in studying circuit function within the neocortex is to understand the spatial and temporal patterns of information flow between different columns and layers. Aim: We aimed to investigate the three-dimensional (3D) layer- and area-specific interactions in mouse cortex in vivo. Approach: We applied a new promising neuroimaging method-fluorescence laminar optical tomography in combination with voltage-sensitive dye imaging (VSDi). VSDi is a powerful technique for interrogating membrane potential dynamics in assemblies of cortical neurons, but it is traditionally used for two-dimensional (2D) imaging. Our mesoscopic technique allows visualization of neuronal activity in a 3D manner with high temporal resolution. Results: We first demonstrated the depth-resolved capability of 3D mesoscopic imaging technology in Thy1-ChR2-YFP transgenic mice. Next, we recorded the long-range functional projections between sensory cortex (S1) and motor cortex (M1) in mice, in vivo, following single whisker deflection. Conclusions: The results show that mesoscopic imaging technique has the potential to investigate the layer-specific neural connectivity in the mouse cortex in vivo. Combination of mesoscopic imaging technique with optogenetic control strategy is a promising platform for determining depth-resolved interactions between cortical circuit elements.
Collapse
Affiliation(s)
- Qinggong Tang
- University of Oklahoma, Stephenson School of Biomedical Engineering, Norman, Oklahoma, United States
- University of Maryland, Fischell Department of Bioengineering, College Park, Maryland, United States
- Address all correspondence to Qinggong Tang, ; Reha S. Erzurumlu, ; Yu Chen,
| | - Vassiliy Tsytsarev
- University of Maryland School of Medicine, Department of Anatomy and Neurobiology, Baltimore, Maryland, United States
| | - Feng Yan
- University of Oklahoma, Stephenson School of Biomedical Engineering, Norman, Oklahoma, United States
| | - Chen Wang
- University of Oklahoma, Stephenson School of Biomedical Engineering, Norman, Oklahoma, United States
| | - Reha S. Erzurumlu
- University of Maryland School of Medicine, Department of Anatomy and Neurobiology, Baltimore, Maryland, United States
- Address all correspondence to Qinggong Tang, ; Reha S. Erzurumlu, ; Yu Chen,
| | - Yu Chen
- University of Maryland, Fischell Department of Bioengineering, College Park, Maryland, United States
- University of Massachusetts, Department of Biomedical Engineering, Amherst, Massachusetts, United States
- Address all correspondence to Qinggong Tang, ; Reha S. Erzurumlu, ; Yu Chen,
| |
Collapse
|
50
|
Hasegawa R, Ebina T, Tanaka YR, Kobayashi K, Matsuzaki M. Structural dynamics and stability of corticocortical and thalamocortical axon terminals during motor learning. PLoS One 2020; 15:e0234930. [PMID: 32559228 PMCID: PMC7304593 DOI: 10.1371/journal.pone.0234930] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/04/2020] [Indexed: 12/30/2022] Open
Abstract
Synaptic plasticity is the cellular basis of learning and memory. When animals learn a novel motor skill, synaptic modifications are induced in the primary motor cortex (M1), and new postsynaptic dendritic spines relevant to motor memory are formed in the early stage of learning. However, it is poorly understood how presynaptic axonal boutons are formed, eliminated, and maintained during motor learning, and whether long-range corticocortical and thalamocortical axonal boutons show distinct structural changes during learning. In this study, we conducted two-photon imaging of presynaptic boutons of long-range axons in layer 1 (L1) of the mouse M1 during the 7-day learning of an accelerating rotarod task. The training-period-averaged rate of formation of boutons on axons projecting from the secondary motor cortical area increased, while the average rate of elimination of those from the motor thalamus (thalamic boutons) decreased. In particular, the elimination rate of thalamic boutons during days 4-7 was lower than that in untrained mice, and the fraction of pre-existing thalamic boutons that survived until day 7 was higher than that in untrained mice. Our results suggest that the late stabilization of thalamic boutons in M1 contributes to motor skill learning.
Collapse
Affiliation(s)
- Ryota Hasegawa
- Division of Brain Circuits, National Institute for Basic Biology, Myodaiji, Okazaki, Japan
- Division of Behavioral Neurobiology, National Institute for Basic Biology, Myodaiji, Okazaki, Japan
- Department of Basic Biology, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Japan
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Teppei Ebina
- Division of Brain Circuits, National Institute for Basic Biology, Myodaiji, Okazaki, Japan
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yasuhiro R. Tanaka
- Division of Brain Circuits, National Institute for Basic Biology, Myodaiji, Okazaki, Japan
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Brain Science Institute, Tamagawa University, Tokyo, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Masanori Matsuzaki
- Division of Brain Circuits, National Institute for Basic Biology, Myodaiji, Okazaki, Japan
- Department of Basic Biology, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Japan
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Brain Functional Dynamics Collaboration Laboratory, RIKEN Center for Brain Science, Saitama, Japan
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo Institutes for Advanced Study, Tokyo, Japan
- * E-mail:
| |
Collapse
|