1
|
Nitzan N, Buzsáki G. Diversity of omission responses to visual images across brain-wide regions. SCIENCE ADVANCES 2025; 11:eadv5651. [PMID: 40397743 DOI: 10.1126/sciadv.adv5651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/17/2025] [Indexed: 05/23/2025]
Abstract
An organism's survival depends on its ability to anticipate forthcoming events and detect discrepancies between the expected and actual sensory inputs. We analyzed data from mice performing a visual go/no-go change-detection task where the sequence of stimulus presentations was intermittently interrupted by omission of a stimulus. The omission of a visual stimulus did not elicit discernable spiking responses in visual cortical neurons. Instead, firing rates between image presentations, including the omission period, ramped linearly and without interruption at the time of the omitted image. Several neuron types in visual cortex neurons were identified with various responses to images and their omissions. A minority of cells in nonvisual areas, including the hippocampus, increased their firing rates at the omitted stimulus onset even when these neurons did not respond to the images. Our study elucidates the origin of omission responses in the visual cortex and sheds light on the role of hippocampal and subcortical circuits in omission detection.
Collapse
Affiliation(s)
- Noam Nitzan
- Neuroscience Institute, New York University, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - György Buzsáki
- Neuroscience Institute, New York University, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| |
Collapse
|
2
|
Yagishita H, Sasaki T. Integrating physiological and transcriptomic analyses at the single-neuron level. Neurosci Res 2025; 214:69-74. [PMID: 38821412 DOI: 10.1016/j.neures.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 04/30/2024] [Accepted: 05/12/2024] [Indexed: 06/02/2024]
Abstract
Neurons generate various spike patterns to execute different functions. Understanding how these physiological neuronal spike patterns are related to their molecular characteristics is a long-standing issue in neuroscience. Herein, we review the results of recent studies that have addressed this issue by integrating physiological and transcriptomic techniques. A sequence of experiments, including in vivo recording and/or labeling, brain tissue slicing, cell collection, and transcriptomic analysis, have identified the gene expression profiles of brain neurons at the single-cell level, with activity patterns recorded in living animals. Although these techniques are still in the early stages, this methodological idea is principally applicable to various brain regions and neuronal activity patterns. Accumulating evidence will contribute to a deeper understanding of neuronal characteristics by integrating insights from molecules to cells, circuits, and behaviors.
Collapse
Affiliation(s)
- Haruya Yagishita
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan.
| |
Collapse
|
3
|
Hui T, Zhou J, Yao M, Xie Y, Zeng H. Advances in Spatial Omics Technologies. SMALL METHODS 2025; 9:e2401171. [PMID: 40099571 DOI: 10.1002/smtd.202401171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/03/2025] [Indexed: 03/20/2025]
Abstract
Rapidly developing spatial omics technologies provide us with new approaches to deeply understanding the diversity and functions of cell types within organisms. Unlike traditional approaches, spatial omics technologies enable researchers to dissect the complex relationships between tissue structure and function at the cellular or even subcellular level. The application of spatial omics technologies provides new perspectives on key biological processes such as nervous system development, organ development, and tumor microenvironment. This review focuses on the advancements and strategies of spatial omics technologies, summarizes their applications in biomedical research, and highlights the power of spatial omics technologies in advancing the understanding of life sciences related to development and disease.
Collapse
Affiliation(s)
- Tianxiao Hui
- State Key Laboratory of Gene Function and Modulation Research, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Jian Zhou
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Muchen Yao
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Yige Xie
- School of Nursing, Peking University, Beijing, 100871, China
| | - Hu Zeng
- State Key Laboratory of Gene Function and Modulation Research, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, 100871, China
| |
Collapse
|
4
|
Malhotra S, Donneger F, Farrell JS, Dudok B, Losonczy A, Soltesz I. Integrating endocannabinoid signaling, CCK interneurons, and hippocampal circuit dynamics in behaving animals. Neuron 2025:S0896-6273(25)00188-6. [PMID: 40267911 DOI: 10.1016/j.neuron.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/25/2025]
Abstract
The brain's endocannabinoid signaling system modulates a diverse range of physiological phenomena and is also involved in various psychiatric and neurological disorders. The basic components of the molecular machinery underlying endocannabinoid-mediated synaptic signaling have been known for decades. However, limitations associated with the short-lived nature of endocannabinoid lipid signals had made it challenging to determine the spatiotemporal specificity and dynamics of endocannabinoid signaling in vivo. Here, we discuss how novel technologies have recently enabled unprecedented insights into endocannabinoid signaling taking place at specific synapses in behaving animals. In this review, we primarily focus on cannabinoid-sensitive inhibition in the hippocampus in relation to place cell properties to illustrate the potential of these novel methodologies. In addition, we highlight implications of these approaches and insights for the unraveling of cannabinoid regulation of synapses in vivo in other brain circuits in both health and disease.
Collapse
Affiliation(s)
- Shreya Malhotra
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
| | - Florian Donneger
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Jordan S Farrell
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Center, Harvard Medical School, Boston, MA, USA
| | - Barna Dudok
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, USA; Kavli Institute for Brain Sciences, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
5
|
Esparza J, Quintanilla JP, Cid E, Medeiros AC, Gallego JA, de la Prida LM. Cell-type-specific manifold analysis discloses independent geometric transformations in the hippocampal spatial code. Neuron 2025; 113:1098-1109.e6. [PMID: 40015277 DOI: 10.1016/j.neuron.2025.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/26/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025]
Abstract
Integrating analyses of genetically defined cell types with population-level approaches remains poorly explored. We investigated this question by focusing on hippocampal spatial maps and the contribution of two genetically defined pyramidal cell types in the deep and superficial CA1 sublayers. Using single- and dual-color miniscope imaging in mice running along a linear track, we found that population activity from these cells exhibited three-dimensional ring manifolds that encoded the animal position and running direction. Despite shared topology, sublayer-specific manifolds displayed distinct geometric features. Manipulating track orientation revealed rotational and translational changes in manifolds from deep cells, contrasting with more stable representations by superficial cells. These transformations were not observed in manifolds derived from the entire CA1 population. Instead, cell-type-specific chemogenetic silencing of either sublayer revealed independent geometric codes. Our results show how genetically specified subpopulations may underpin parallel spatial maps that can be manipulated independently.
Collapse
Affiliation(s)
| | | | - Elena Cid
- Instituto Cajal CSIC, Madrid 28002, Spain
| | - Ana C Medeiros
- Instituto Cajal CSIC, Madrid 28002, Spain; Faculdade de Medicina de Riberâo Preto, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Juan A Gallego
- Department of Bioengineering, Imperial College London, London, UK
| | | |
Collapse
|
6
|
Arkhipov A, da Costa N, de Vries S, Bakken T, Bennett C, Bernard A, Berg J, Buice M, Collman F, Daigle T, Garrett M, Gouwens N, Groblewski PA, Harris J, Hawrylycz M, Hodge R, Jarsky T, Kalmbach B, Lecoq J, Lee B, Lein E, Levi B, Mihalas S, Ng L, Olsen S, Reid C, Siegle JH, Sorensen S, Tasic B, Thompson C, Ting JT, van Velthoven C, Yao S, Yao Z, Koch C, Zeng H. Integrating multimodal data to understand cortical circuit architecture and function. Nat Neurosci 2025; 28:717-730. [PMID: 40128391 DOI: 10.1038/s41593-025-01904-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/21/2025] [Indexed: 03/26/2025]
Abstract
In recent years there has been a tremendous growth in new technologies that allow large-scale investigation of different characteristics of the nervous system at an unprecedented level of detail. There is a growing trend to use combinations of these new techniques to determine direct links between different modalities. In this Perspective, we focus on the mouse visual cortex, as this is one of the model systems in which much progress has been made in the integration of multimodal data to advance understanding. We review several approaches that allow integration of data regarding various properties of cortical cell types, connectivity at the level of brain areas, cell types and individual cells, and functional neural activity in vivo. The increasingly crucial contributions of computation and theory in analyzing and systematically modeling data are also highlighted. Together with open sharing of data, tools and models, integrative approaches are essential tools in modern neuroscience for improving our understanding of the brain architecture, mechanisms and function.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jim Berg
- Allen Institute, Seattle, WA, USA
| | | | | | | | | | | | | | - Julie Harris
- Allen Institute, Seattle, WA, USA
- Cure Alzheimer's Fund, Wellesley Hills, MA, USA
| | | | | | | | | | | | | | - Ed Lein
- Allen Institute, Seattle, WA, USA
| | | | | | - Lydia Ng
- Allen Institute, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Schneider-Mizell CM, Bodor AL, Brittain D, Buchanan J, Bumbarger DJ, Elabbady L, Gamlin C, Kapner D, Kinn S, Mahalingam G, Seshamani S, Suckow S, Takeno M, Torres R, Yin W, Dorkenwald S, Bae JA, Castro MA, Halageri A, Jia Z, Jordan C, Kemnitz N, Lee K, Li K, Lu R, Macrina T, Mitchell E, Mondal SS, Mu S, Nehoran B, Popovych S, Silversmith W, Turner NL, Wong W, Wu J, Reimer J, Tolias AS, Seung HS, Reid RC, Collman F, da Costa NM. Inhibitory specificity from a connectomic census of mouse visual cortex. Nature 2025; 640:448-458. [PMID: 40205209 PMCID: PMC11981935 DOI: 10.1038/s41586-024-07780-8] [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] [Received: 03/27/2023] [Accepted: 07/03/2024] [Indexed: 04/11/2025]
Abstract
Mammalian cortex features a vast diversity of neuronal cell types, each with characteristic anatomical, molecular and functional properties1. Synaptic connectivity shapes how each cell type participates in the cortical circuit, but mapping connectivity rules at the resolution of distinct cell types remains difficult. Here we used millimetre-scale volumetric electron microscopy2 to investigate the connectivity of all inhibitory neurons across a densely segmented neuronal population of 1,352 cells spanning all layers of mouse visual cortex, producing a wiring diagram of inhibition with more than 70,000 synapses. Inspired by classical neuroanatomy, we classified inhibitory neurons based on targeting of dendritic compartments and developed an excitatory neuron classification based on dendritic reconstructions with whole-cell maps of synaptic input. Single-cell connectivity showed a class of disinhibitory specialist that targets basket cells. Analysis of inhibitory connectivity onto excitatory neurons found widespread specificity, with many interneurons exhibiting differential targeting of spatially intermingled subpopulations. Inhibitory targeting was organized into 'motif groups', diverse sets of cells that collectively target both perisomatic and dendritic compartments of the same excitatory targets. Collectively, our analysis identified new organizing principles for cortical inhibition and will serve as a foundation for linking contemporary multimodal neuronal atlases with the cortical wiring diagram.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Clare Gamlin
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Sam Kinn
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Marc Takeno
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Wenjing Yin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Sven Dorkenwald
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - J Alexander Bae
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Electrical and Computer Engineering Department, Princeton University, Princeton, NJ, USA
| | - Manuel A Castro
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Akhilesh Halageri
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Zhen Jia
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - Chris Jordan
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Nico Kemnitz
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Kisuk Lee
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Kai Li
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - Ran Lu
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Thomas Macrina
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - Eric Mitchell
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Shanka Subhra Mondal
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Electrical and Computer Engineering Department, Princeton University, Princeton, NJ, USA
| | - Shang Mu
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Barak Nehoran
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - Sergiy Popovych
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | | | - Nicholas L Turner
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - William Wong
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Jingpeng Wu
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Jacob Reimer
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
| | - Andreas S Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Center for Neuroscience and Artificial Intelligence, Baylor College of Medicine, Houston, TX, USA
- Brain & Cognitive Sciences Department, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - H Sebastian Seung
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - R Clay Reid
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | |
Collapse
|
8
|
Poirier J, Beninger J, Naud R. Computational protocol for modeling and analyzing synaptic dynamics using SRPlasticity. STAR Protoc 2025; 6:103652. [PMID: 40029747 PMCID: PMC11915160 DOI: 10.1016/j.xpro.2025.103652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/09/2024] [Accepted: 02/05/2025] [Indexed: 03/21/2025] Open
Abstract
Transient changes in synaptic strength, known as short-term plasticity (STP), play a fundamental role in neuronal communication. Here, we present a protocol for using SRPlasticity, a software package that implements a computational model of STP. SRPlasticity supports automatic characterization of electrophysiological data and simulation of synaptic responses. We describe steps for installing and utilizing SRPlasticity, preprocessing data, fitting models, and simulating responses. We then detail procedures for analyzing spike response plasticity (SRP) model parameters to infer functional groupings of STP. For complete details on the use and execution of this protocol, please refer to Rossbroich et al.1 and Beninger et al.2.
Collapse
Affiliation(s)
- Jade Poirier
- Center for Neural Dynamics and Artificial Intelligence, uOttawa Brain and Mind Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - John Beninger
- Center for Neural Dynamics and Artificial Intelligence, uOttawa Brain and Mind Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| | - Richard Naud
- Center for Neural Dynamics and Artificial Intelligence, uOttawa Brain and Mind Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Physics, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
9
|
Meier AM, D'Souza RD, Ji W, Han EB, Burkhalter A. Interdigitating Modules for Visual Processing During Locomotion and Rest in Mouse V1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639505. [PMID: 40060542 PMCID: PMC11888233 DOI: 10.1101/2025.02.21.639505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Layer 1 of V1 has been shown to receive locomotion-related signals from the dorsal lateral geniculate (dLGN) and lateral posterior (LP) thalamic nuclei (Roth et al., 2016). Inputs from the dLGN terminate in M2+ patches while inputs from LP target M2- interpatches (D'Souza et al., 2019) suggesting that motion related signals are processed in distinct networks. Here, we investigated by calcium imaging in head-fixed awake mice whether L2/3 neurons underneath L1 M2+ and M2- modules are differentially activated by locomotion, and whether distinct networks of feedback connections from higher cortical areas to L1 may contribute to these differences. We found that strongly locomotion-modulated cell clusters during visual stimulation were aligned with M2- interpatches, while weakly modulated cells clustered under M2+ patches. Unlike M2+ patch cells, pairs of M2- interpatch cells showed increased correlated variability of calcium transients when the sites in the visuotopic map were far apart, suggesting that activity is integrated across large parts of the visual field. Pathway tracing further suggests that strong locomotion modulation in L2/3 M2- interpatch cells of V1 relies on looped, like-to-like networks between apical dendrites of MOs-, PM- and RSP-projecting neurons and feedback input from these areas to L1. M2- interpatches receive strong inputs from SST neurons, suggesting that during locomotion these interneurons influence the firing of specific subnetworks by controlling the excitability of apical dendrites in M2- interpatches.
Collapse
Affiliation(s)
- A M Meier
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110; USA
| | - R D D'Souza
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110; USA
| | - W Ji
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110; USA
| | - E B Han
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110; USA
| | - A Burkhalter
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110; USA
| |
Collapse
|
10
|
English D, Gilbert E, Klaver L, Arndt K, Kim J, Jia X, Mckenzie S. Reciprocal interactions between CA1 pyramidal and axo-axonic cells control sharp wave-ripple events. RESEARCH SQUARE 2025:rs.3.rs-5844238. [PMID: 39989976 PMCID: PMC11844635 DOI: 10.21203/rs.3.rs-5844238/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Diverse sources of inhibition serve to modulate circuits and control cell assembly spiking across various timescales. For example, in hippocampus area CA1 the competition between inhibition and excitation organizes spike timing of pyramidal cells (PYR) in network events, including sharp wave-ripples (SPW-R). Specific cellular-synaptic sources of inhibition in SPW-R remain unclear, as there are > 20 types of GABAergic interneurons in CA1. Axo-axonic cells (AAC) are defined by their synaptic targeting of the axon initial segment of pyramidal cells, potently controlling spike output. The impact of AAC activity on SPW-R is controversial, due mainly to ambiguity of AAC identification. Here we monitored and manipulated opto-tagged AACs in behaving mice using silicon probe recordings. We found a large variability of AAC neurons, varying from enhanced to suppressed spiking during SPW-Rs, in contrast to the near-uniform excitation of other parvalbumin-expressing interneurons. AACs received convergent monosynaptic inputs from local pyramidal cell assemblies, which strongly influenced their participation in SPW-Rs. Optogenetic silencing of AACs increased power and duration of SPW-Rs, recruiting a greater number of PYR, suggesting AACs control SPW-R dynamics. We hypothesize that lateral inhibition by reciprocal PYR-AAC interactions thus supports the organization of cell assemblies in SPW-R.
Collapse
Affiliation(s)
| | - Earl Gilbert
- Virginia Polytechnic Institute and State University
| | | | | | | | - Xiaoting Jia
- Virginia Polytechnic Institute and State University
| | | |
Collapse
|
11
|
Posani L, Wang S, Muscinelli SP, Paninski L, Fusi S. Rarely categorical, always high-dimensional: how the neural code changes along the cortical hierarchy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.15.623878. [PMID: 39605683 PMCID: PMC11601379 DOI: 10.1101/2024.11.15.623878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
A long-standing debate in neuroscience concerns whether individual neurons are organized into functionally distinct populations that encode information differently ("categorical" representations [1-3]) and the implications for neural computation. Here, we systematically analyzed how cortical neurons encode cognitive, sensory, and movement variables across 43 cortical regions during a complex task (14,000+ units from the International Brain Laboratory public Brain-wide Map data set [4]) and studied how these properties change across the sensory-cognitive cortical hierarchy [5]. We found that the structure of the neural code was scale-dependent: on a whole-cortex scale, neural selectivity was categorical and organized across regions in a way that reflected their anatomical connectivity. However, within individual regions, categorical representations were rare and limited to primary sensory areas. Remarkably, the degree of categorical clustering of neural selectivity was inversely correlated to the dimensionality of neural representations, suggesting a link between single-neuron selectivity and computational properties of population codes that we explained in a mathematical model. Finally, we found that the fraction of linearly separable combinations of experimental conditions ("Shattering Dimensionality" [6]) was near maximal across all areas, indicating a robust and uniform ability for flexible information encoding throughout the cortex. In conclusion, our results provide systematic evidence for a non-categorical, high-dimensional neural code in all but the lower levels of the cortical hierarchy.
Collapse
Affiliation(s)
- Lorenzo Posani
- Zuckerman Institute, Columbia University, New York, NY, USA
- School of Computer and Communication Sciences, EPFL, Street, Lausanne, Switzerland
| | - Shuqi Wang
- School of Computer and Communication Sciences, EPFL, Street, Lausanne, Switzerland
- Department of Statistics, Columbia University, New York, NY, USA
| | | | - Liam Paninski
- Zuckerman Institute, Columbia University, New York, NY, USA
- Department of Statistics, Columbia University, New York, NY, USA
- Co-senior authors
| | - Stefano Fusi
- Zuckerman Institute, Columbia University, New York, NY, USA
- Co-senior authors
| |
Collapse
|
12
|
Turner KL, Brockway DF, Hossain MS, Griffith KR, Greenawalt DI, Zhang Q, Gheres KW, Crowley NA, Drew PJ. Type-I nNOS neurons orchestrate cortical neural activity and vasomotion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634042. [PMID: 39896560 PMCID: PMC11785022 DOI: 10.1101/2025.01.21.634042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
It is unknown how the brain orchestrates coordination of global neural and vascular dynamics. We sought to uncover the role of a sparse but unusual population of genetically-distinct interneurons known as type-I nNOS neurons, using a novel pharmacological strategic to unilaterally ablate these neurons from the somatosensory cortex of mice. Region-specific ablation produced changes in both neural activity and vascular dynamics, decreased power in the delta-band of the local field potential, reduced sustained vascular responses to prolonged sensory stimulation, and abolished the post-stimulus undershoot in cerebral blood volume. Coherence between the left and right somatosensory cortex gamma-band power envelope and blood volume at ultra-low frequencies was decreased, suggesting type-1 nNOS neurons integrate long-range coordination of brain signals. Lastly, we observed decreases in the amplitude of resting-state blood volume oscillations and decreased vasomotion following the ablation of type-I nNOS neurons. This demonstrates that a small population of nNOS-positive neurons are indispensable for regulating both neural and vascular dynamics in the whole brain and implicates disruption of these neurons in diseases ranging from neurodegeneration to sleep disturbances.
Collapse
Affiliation(s)
- Kevin L. Turner
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Dakota F. Brockway
- Department of Biology, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Md Shakhawat Hossain
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Keith R. Griffith
- Department of Biology, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Denver I. Greenawalt
- Graduate Program in Molecular Cellular and Integrative Biosciences, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Qingguang Zhang
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
- Department of Physiology, Michigan State University, East Lansing, MI 48824
| | - Kyle W. Gheres
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Nicole A. Crowley
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Biology, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| | - Patrick J. Drew
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802
- Department of Neurosurgery, The Pennsylvania State University, University Park, PA 16802
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
13
|
Hozhabri E, Corredera Asensio A, Elmaleh M, Kim JW, Phillips MB, Frazel PW, Dimidschstein J, Fishell G, Long MA. Differential behavioral engagement of inhibitory interneuron subtypes in the zebra finch brain. Neuron 2025; 113:460-470.e7. [PMID: 39644901 PMCID: PMC11802303 DOI: 10.1016/j.neuron.2024.11.003] [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: 05/18/2024] [Revised: 08/30/2024] [Accepted: 11/07/2024] [Indexed: 12/09/2024]
Abstract
Inhibitory interneurons are highly heterogeneous circuit elements often characterized by cell biological properties, but how these factors relate to specific roles underlying complex behavior remains poorly understood. Using chronic silicon probe recordings, we demonstrate that distinct interneuron groups perform different inhibitory roles within HVC, a song production circuit in the zebra finch forebrain. To link these functional subtypes to molecular identity, we performed two-photon targeted electrophysiological recordings of HVC interneurons followed by post hoc immunohistochemistry of subtype-specific markers. We find that parvalbumin-expressing interneurons are highly modulated by sensory input and likely mediate auditory gating, whereas a more heterogeneous set of somatostatin-expressing interneurons can strongly regulate activity based on arousal. Using this strategy, we uncover important cell-type-specific network functions in the context of an ethologically relevant motor skill.
Collapse
Affiliation(s)
- Ellie Hozhabri
- NYU Neuroscience Institute and Department of Otolaryngology, New York University Langone Medical Center, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA
| | - Ariadna Corredera Asensio
- NYU Neuroscience Institute and Department of Otolaryngology, New York University Langone Medical Center, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA
| | - Margot Elmaleh
- NYU Neuroscience Institute and Department of Otolaryngology, New York University Langone Medical Center, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA
| | - Jeong Woo Kim
- NYU Neuroscience Institute and Department of Otolaryngology, New York University Langone Medical Center, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA
| | - Matthew B Phillips
- NYU Neuroscience Institute and Department of Otolaryngology, New York University Langone Medical Center, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA
| | - Paul W Frazel
- NYU Neuroscience Institute and Department of Otolaryngology, New York University Langone Medical Center, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Gord Fishell
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Michael A Long
- NYU Neuroscience Institute and Department of Otolaryngology, New York University Langone Medical Center, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
14
|
Shainer I, Kappel JM, Laurell E, Donovan JC, Schneider MW, Kuehn E, Arnold-Ammer I, Stemmer M, Larsch J, Baier H. Transcriptomic neuron types vary topographically in function and morphology. Nature 2025; 638:1023-1033. [PMID: 39939759 PMCID: PMC11864986 DOI: 10.1038/s41586-024-08518-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 12/11/2024] [Indexed: 02/14/2025]
Abstract
Neuronal phenotypic traits such as morphology, connectivity and function are dictated, to a large extent, by a specific combination of differentially expressed genes. Clusters of neurons in transcriptomic space correspond to distinct cell types and in some cases-for example, Caenorhabditis elegans neurons1 and retinal ganglion cells2-4-have been shown to share morphology and function. The zebrafish optic tectum is composed of a spatial array of neurons that transforms visual inputs into motor outputs. Although the visuotopic map is continuous, subregions of the tectum are functionally specialized5,6. Here, to uncover the cell-type architecture of the tectum, we transcriptionally profiled its neurons, revealing more than 60 cell types that are organized in distinct anatomical layers. We measured the visual responses of thousands of tectal neurons by two-photon calcium imaging and matched them with their transcriptional profiles. Furthermore, we characterized the morphologies of transcriptionally identified neurons using specific transgenic lines. Notably, we found that neurons that are transcriptionally similar can diverge in shape, connectivity and visual responses. Incorporating the spatial coordinates of neurons within the tectal volume revealed functionally and morphologically defined anatomical subclusters within individual transcriptomic clusters. Our findings demonstrate that extrinsic, position-dependent factors expand the phenotypic repertoire of genetically similar neurons.
Collapse
Affiliation(s)
- Inbal Shainer
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Johannes M Kappel
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Eva Laurell
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Joseph C Donovan
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | | | - Enrico Kuehn
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | | | - Manuel Stemmer
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Johannes Larsch
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Herwig Baier
- Max Planck Institute for Biological Intelligence, Martinsried, Germany.
| |
Collapse
|
15
|
Park E, Mosso MB, Barth AL. Neocortical somatostatin neuron diversity in cognition and learning. Trends Neurosci 2025; 48:140-155. [PMID: 39824710 DOI: 10.1016/j.tins.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/13/2024] [Accepted: 12/12/2024] [Indexed: 01/20/2025]
Abstract
Somatostatin-expressing (SST) neurons are a major class of electrophysiologically and morphologically distinct inhibitory cells in the mammalian neocortex. Transcriptomic data suggest that this class can be divided into multiple subtypes that are correlated with morpho-electric properties. At the same time, availability of transgenic tools to identify and record from SST neurons in awake, behaving mice has stimulated insights about their response properties and computational function. Neocortical SST neurons are regulated by sleep and arousal, attention, and novelty detection, and show marked response plasticity during learning. Recent studies suggest that subtype-specific analysis of SST neurons may be critical for understanding their complex roles in cortical function. In this review, we discuss and synthesize recent advances in understanding the diversity, circuit integration, and functional properties of this important group of GABAergic neurons.
Collapse
Affiliation(s)
- Eunsol Park
- Department of Biological Sciences and Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Matthew B Mosso
- Department of Biological Sciences and Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Alison L Barth
- Department of Biological Sciences and Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
McLachlan CA, Lee DG, Kwon O, Delgado KM, Manjrekar N, Yao Z, Zeng H, Tasic B, Chen JL. Transcriptional determinants of goal-directed learning and representational drift in the parahippocampal cortex. Cell Rep 2025; 44:115175. [PMID: 39792551 PMCID: PMC11920904 DOI: 10.1016/j.celrep.2024.115175] [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: 02/25/2024] [Revised: 10/21/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
Task learning involves learning associations between stimuli and outcomes and storing these relationships in memory. While this information can be reliably decoded from population activity, individual neurons encoding this representation can drift over time. The circuit or molecular mechanisms underlying this drift and its role in learning are unclear. We performed two-photon calcium imaging in the perirhinal cortex during task training. Using post hoc spatial transcriptomics, we measured immediate-early gene (IEG) expression and assigned monitored neurons to excitatory or inhibitory subtypes. We discovered an IEG-defined network spanning multiple subtypes that form stimulus-outcome associations. Targeted deletion of brain-derived neurotrophic factor in the perirhinal cortex disrupted IEG expression and impaired task learning. Representational drift slowed with prolonged training. Pre-existing representations were strengthened while stimulus-reward associations failed to form. Our findings reveal the cell types and molecules regulating long-term network stability that is permissive for task learning and memory allocation.
Collapse
Affiliation(s)
- Caroline A McLachlan
- Department of Biology, Boston University, Boston, MA 02215, USA; Center for Neurophotonics, Boston University, Boston, MA 02215, USA
| | - David G Lee
- Center for Neurophotonics, Boston University, Boston, MA 02215, USA; Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Osung Kwon
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Kevin M Delgado
- Department of Biology, Boston University, Boston, MA 02215, USA
| | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jerry L Chen
- Department of Biology, Boston University, Boston, MA 02215, USA; Center for Neurophotonics, Boston University, Boston, MA 02215, USA; Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Center for Systems Neuroscience, Boston University, Boston MA 02215, USA.
| |
Collapse
|
17
|
Gilbert ET, Klaver LMF, Arndt KC, Kim J, Jia X, McKenzie S, English DF. Reciprocal interactions between CA1 pyramidal and axo-axonic cells control sharp wave-ripple events. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.02.601726. [PMID: 39868302 PMCID: PMC11761640 DOI: 10.1101/2024.07.02.601726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Diverse sources of inhibition serve to modulate circuits and control cell assembly spiking across various timescales. For example, in hippocampus area CA1 the competition between inhibition and excitation organizes spike timing of pyramidal cells (PYR) in network events, including sharp wave-ripples (SPW-R). Specific cellular-synaptic sources of inhibition in SPW-R remain unclear, as there are >20 types of GABAergic interneurons in CA1. Axo-axonic cells (AAC) are defined by their synaptic targeting of the axon initial segment of pyramidal cells, potently controlling spike output. The impact of AAC activity on SPW-R is controversial, due mainly to ambiguity of AAC identification. Here we monitored and manipulated opto-tagged AACs in behaving mice using silicon probe recordings. We found a large variability of AAC neurons, varying from enhanced to suppressed spiking during SPW-Rs, in contrast to the near-uniform excitation of other parvalbumin-expressing interneurons. AACs received convergent monosynaptic inputs from local pyramidal cell assemblies, which strongly influenced their participation in SPW-Rs. Optogenetic silencing of AACs increased power and duration of SPW-Rs, recruiting a greater number of PYR, suggesting AACs control SPW-R dynamics. We hypothesize that lateral inhibition by reciprocal PYR-AAC interactions thus supports the organization of cell assemblies in SPW-R.
Collapse
|
18
|
Kaplan HS, Horvath PM, Rahman MM, Dulac C. The neurobiology of parenting and infant-evoked aggression. Physiol Rev 2025; 105:315-381. [PMID: 39146250 DOI: 10.1152/physrev.00036.2023] [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: 09/21/2023] [Revised: 07/19/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Parenting behavior comprises a variety of adult-infant and adult-adult interactions across multiple timescales. The state transition from nonparent to parent requires an extensive reorganization of individual priorities and physiology and is facilitated by combinatorial hormone action on specific cell types that are integrated throughout interconnected and brainwide neuronal circuits. In this review, we take a comprehensive approach to integrate historical and current literature on each of these topics across multiple species, with a focus on rodents. New and emerging molecular, circuit-based, and computational technologies have recently been used to address outstanding gaps in our current framework of knowledge on infant-directed behavior. This work is raising fundamental questions about the interplay between instinctive and learned components of parenting and the mutual regulation of affiliative versus agonistic infant-directed behaviors in health and disease. Whenever possible, we point to how these technologies have helped gain novel insights and opened new avenues of research into the neurobiology of parenting. We hope this review will serve as an introduction for those new to the field, a comprehensive resource for those already studying parenting, and a guidepost for designing future studies.
Collapse
Affiliation(s)
- Harris S Kaplan
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Patricia M Horvath
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Mohammed Mostafizur Rahman
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Catherine Dulac
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| |
Collapse
|
19
|
Li Z, He L, Peng L, Zhu X, Li M, Hu D. Negative hemodynamic response in the visual cortex: Evidence supporting neuronal origin via hemodynamic observation and two-photon imaging. Brain Res Bull 2025; 220:111149. [PMID: 39615859 DOI: 10.1016/j.brainresbull.2024.111149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 11/07/2024] [Accepted: 11/25/2024] [Indexed: 12/08/2024]
Abstract
The positive hemodynamic response (PHR) during stimulation often co-occurs with a strong, sustained negative hemodynamic response (NHR). However, the characteristics and neurophysiological mechanisms of the NHR, especially in regions distal to the PHR, remain incompletely understood. Using intrinsic optical imaging (OI) and two-photon imaging, we observed that forelimb electrical stimulation evoked strong PHR signals in the forelimb region of the primary somatosensory cortex (S1FL). Meanwhile, NHR signals primarily appeared in the primary visual cortex (V1), with a delayed onset and lower amplitude relative to the PHR signals. Additionally, stimulation led to a reduction in cerebral blood flow (CBF) in the NHR region. Notably, there was an overall suppression of the calcium response in the NHR region, although a small proportion (14 %) of neurons exhibited concurrent activation. Axon tracing revealed cortico-cortical projections from S1FL to V1. These findings suggest that neuronal deactivation significantly contributes to the origin of the NHR, offering additional insights into the specific inhibitory mechanisms underlying the NHR.
Collapse
Affiliation(s)
- Zhen Li
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Lihua He
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Limin Peng
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Xuan Zhu
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China
| | - Ming Li
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China.
| | - Dewen Hu
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, China.
| |
Collapse
|
20
|
Gonzalez J, Torterolo P, Bolding KA, Tort AB. Communication subspace dynamics of the canonical olfactory pathway. iScience 2024; 27:111275. [PMID: 39628563 PMCID: PMC11613203 DOI: 10.1016/j.isci.2024.111275] [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: 06/07/2024] [Revised: 08/08/2024] [Accepted: 10/25/2024] [Indexed: 12/06/2024] Open
Abstract
Understanding how different brain areas communicate is crucial for elucidating the mechanisms underlying cognition. A possible way for neural populations to interact is through a communication subspace, a specific region in the state-space enabling the transmission of behaviorally relevant spiking patterns. In the olfactory system, it remains unclear if different populations employ such a mechanism. Our study reveals that neuronal ensembles in the main olfactory pathway (olfactory bulb to olfactory cortex) interact through a communication subspace, which is driven by nasal respiration and allows feedforward and feedback transmission to occur segregated along the sniffing cycle. Moreover, our results demonstrate that subspace communication depends causally on the activity of both areas, is hindered during anesthesia, and transmits a low-dimensional representation of odor.
Collapse
Affiliation(s)
- Joaquín Gonzalez
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo 11200, Uruguay
- Brain Institute, Federal University of Rio Grande do Norte, Natal, RN 59078, Brazil
| | - Pablo Torterolo
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo 11200, Uruguay
| | | | - Adriano B.L. Tort
- Brain Institute, Federal University of Rio Grande do Norte, Natal, RN 59078, Brazil
| |
Collapse
|
21
|
Martin N, Olsen P, Quon J, Campos J, Cuevas NV, Nagra J, VanNess M, Maltzer Z, Gelfand EC, Oyama A, Gary A, Wang Y, Alaya A, Ruiz A, Reynoldson C, Bielstein C, Pom CA, Huang C, Slaughterbeck C, Liang E, Alexander J, Ariza J, Malone J, Melchor J, Colbert K, Brouner K, Shulga L, Reding M, Latimer P, Sanchez R, Barta S, Egdorf T, Madigan Z, Pagan CM, Close JL, Long B, Kunst M, Lein ES, Zeng H, McMillen D, Waters J. MerQuaCo: a computational tool for quality control in image-based spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626766. [PMID: 39677693 PMCID: PMC11643037 DOI: 10.1101/2024.12.04.626766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Image-based spatial transcriptomics platforms are powerful tools often used to identify cell populations and describe gene expression in intact tissue. Spatial experiments return large, high-dimension datasets and several open-source software packages are available to facilitate analysis and visualization. Spatial results are typically imperfect. For example, local variations in transcript detection probability are common. Software tools to characterize imperfections and their impact on downstream analyses are lacking so the data quality is assessed manually, a laborious and often a subjective process. Here we describe imperfections in a dataset of 641 fresh-frozen adult mouse brain sections collected using the Vizgen MERSCOPE. Common imperfections included the local loss of tissue from the section, tissue outside the imaging volume due to detachment from the coverslip, transcripts missing due to dropped images, varying detection probability through space, and differences in transcript detection probability between experiments. We describe the incidence of each imperfection and the likely impact on the accuracy of cell type labels. We develop MerQuaCo, open-source code that detects and quantifies imperfections without user input, facilitating the selection of sections for further analysis with existing packages. Together, our results and MerQuaCo facilitate rigorous, objective assessment of the quality of spatial transcriptomics results.
Collapse
Affiliation(s)
| | | | - Jacob Quon
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jazmin Campos
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | | | - Josh Nagra
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Marshall VanNess
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Zoe Maltzer
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Emily C Gelfand
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Alana Oyama
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Amanda Gary
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Yimin Wang
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Angela Alaya
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Augustin Ruiz
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Cade Reynoldson
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | | | | | - Cindy Huang
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | | | - Elizabeth Liang
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jason Alexander
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jeanelle Ariza
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jocelin Malone
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jose Melchor
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Kaity Colbert
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Krissy Brouner
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Lyudmila Shulga
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Melissa Reding
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Patrick Latimer
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Raymond Sanchez
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Stuard Barta
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Tom Egdorf
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Zachary Madigan
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Chelsea M Pagan
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jennie L Close
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Brian Long
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Michael Kunst
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Ed S Lein
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Hongkui Zeng
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Delissa McMillen
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jack Waters
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| |
Collapse
|
22
|
Patiño M, Rossa MA, Lagos WN, Patne NS, Callaway EM. Transcriptomic cell-type specificity of local cortical circuits. Neuron 2024; 112:3851-3866.e4. [PMID: 39353431 PMCID: PMC11624072 DOI: 10.1016/j.neuron.2024.09.003] [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: 02/28/2024] [Revised: 07/02/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
Complex neocortical functions rely on networks of diverse excitatory and inhibitory neurons. While local connectivity rules between major neuronal subclasses have been established, the specificity of connections at the level of transcriptomic subtypes remains unclear. We introduce single transcriptome assisted rabies tracing (START), a method combining monosynaptic rabies tracing and single-nuclei RNA sequencing to identify transcriptomic cell types, providing inputs to defined neuron populations. We employ START to transcriptomically characterize inhibitory neurons providing monosynaptic input to 5 different layer-specific excitatory cortical neuron populations in mouse primary visual cortex (V1). At the subclass level, we observe results consistent with findings from prior studies that resolve neuronal subclasses using antibody staining, transgenic mouse lines, and morphological reconstruction. With improved neuronal subtype granularity achieved with START, we demonstrate transcriptomic subtype specificity of inhibitory inputs to various excitatory neuron subclasses. These results establish local connectivity rules at the resolution of transcriptomic inhibitory cell types.
Collapse
Affiliation(s)
- Maribel Patiño
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA; Medical Scientist Training Program, University of California, San Diego, La Jolla, CA, USA
| | - Marley A Rossa
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Willian Nuñez Lagos
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Neelakshi S Patne
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neuroscience Graduate Program, Boston University, Boston, MA, USA
| | - Edward M Callaway
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
23
|
Colonna M, Konopka G, Liddelow SA, Nowakowski T, Awatramani R, Bateup HS, Cadwell CR, Caglayan E, Chen JL, Gillis J, Kampmann M, Krienen F, Marsh SE, Monje M, O'Dea MR, Patani R, Pollen AA, Quintana FJ, Scavuzzo M, Schmitz M, Sloan SA, Tesar PJ, Tollkuhn J, Tosches MA, Urbanek ME, Werner JM, Bayraktar OA, Gokce O, Habib N. Implementation and validation of single-cell genomics experiments in neuroscience. Nat Neurosci 2024; 27:2310-2325. [PMID: 39627589 DOI: 10.1038/s41593-024-01814-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 10/15/2024] [Indexed: 12/13/2024]
Abstract
Single-cell or single-nucleus transcriptomics is a powerful tool for identifying cell types and cell states. However, hypotheses derived from these assays, including gene expression information, require validation, and their functional relevance needs to be established. The choice of validation depends on numerous factors. Here, we present types of orthogonal and functional validation experiment to strengthen preliminary findings obtained using single-cell and single-nucleus transcriptomics as well as the challenges and limitations of these approaches.
Collapse
Affiliation(s)
- Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| | - Genevieve Konopka
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| | - Tomasz Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
| | - Rajeshwar Awatramani
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, USA
| | - Helen S Bateup
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Cathryn R Cadwell
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Emre Caglayan
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jerry L Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Center for Neurophotonics, Boston University, Boston, MA, USA
- Department of Biology, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Jesse Gillis
- Department of Physiology and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Fenna Krienen
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Samuel E Marsh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Michael R O'Dea
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Rickie Patani
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, Human Stem Cells and Neurodegeneration Laboratory, London, UK
| | - Alex A Pollen
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marissa Scavuzzo
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, OH, USA
- Institute for Glial Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Matthew Schmitz
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, OH, USA
- Institute for Glial Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | - Madeleine E Urbanek
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan M Werner
- Department of Physiology and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Ozgun Gokce
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, Bonn, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| | - Naomi Habib
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
24
|
Yu H, Lyu H, Xu EY, Windolf C, Lee EK, Yang F, Shelton AM, Olsen S, Minavi S, Winter O, Dyer EL, Chandrasekaran C, Steinmetz NA, Paninski L, Hurwitz C. IN VIVO CELL-TYPE AND BRAIN REGION CLASSIFICATION VIA MULTIMODAL CONTRASTIVE LEARNING. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.05.622159. [PMID: 39574717 PMCID: PMC11580900 DOI: 10.1101/2024.11.05.622159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Current electrophysiological approaches can track the activity of many neurons, yet it is usually unknown which cell-types or brain areas are being recorded without further molecular or histological analysis. Developing accurate and scalable algorithms for identifying the cell-type and brain region of recorded neurons is thus crucial for improving our understanding of neural computation. In this work, we develop a multimodal contrastive learning approach for neural data that can be fine-tuned for different downstream tasks, including inference of cell-type and brain location. We utilize this approach to jointly embed the activity autocorrelations and extracellular waveforms of individual neurons. We demonstrate that our embedding approach, Neuronal Embeddings via MultimOdal contrastive learning (NEMO), paired with supervised fine-tuning, achieves state-of-the-art cell-type classification for an opto-tagged visual cortex dataset and brain region classification for the public International Brain Laboratory brain-wide map dataset. Our method represents a promising step towards accurate cell-type and brain region classification from electrophysiological recordings.
Collapse
Affiliation(s)
- Han Yu
- Columbia University, New York, NY, USA
| | - Hanrui Lyu
- Northwestern University, Evanston, IL, USA
| | | | | | | | - Fan Yang
- University College London, London, UK
| | | | | | | | | | - Eva L Dyer
- Georgia Institute of Technology, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
25
|
Cang J, Chen C, Li C, Liu Y. Genetically defined neuron types underlying visuomotor transformation in the superior colliculus. Nat Rev Neurosci 2024; 25:726-739. [PMID: 39333418 DOI: 10.1038/s41583-024-00856-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2024] [Indexed: 09/29/2024]
Abstract
The superior colliculus (SC) is a conserved midbrain structure that is important for transforming visual and other sensory information into motor actions. Decades of investigations in numerous species have made the SC and its nonmammalian homologue, the optic tectum, one of the best studied structures in the brain, with rich information now available regarding its anatomical organization, its extensive inputs and outputs and its important functions in many reflexive and cognitive behaviours. Excitingly, recent studies using modern genomic and physiological approaches have begun to reveal the diverse neuronal subtypes in the SC, as well as their unique functions in visuomotor transformation. Studies have also started to uncover how subtypes of SC neurons form intricate circuits to mediate visual processing and visually guided behaviours. Here, we review these recent discoveries on the cell types and neuronal circuits underlying visuomotor transformations mediated by the SC. We also highlight the important future directions made possible by these new developments.
Collapse
Affiliation(s)
- Jianhua Cang
- Department of Biology, University of Virginia, Charlottesville, VA, USA.
- Department of Psychology, University of Virginia, Charlottesville, VA, USA.
| | - Chen Chen
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Chuiwen Li
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Yuanming Liu
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
26
|
Chen X. Reimagining Cortical Connectivity by Deconstructing Its Molecular Logic into Building Blocks. Cold Spring Harb Perspect Biol 2024; 16:a041509. [PMID: 38621822 PMCID: PMC11529856 DOI: 10.1101/cshperspect.a041509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Comprehensive maps of neuronal connectivity provide a foundation for understanding the structure of neural circuits. In a circuit, neurons are diverse in morphology, electrophysiology, gene expression, activity, and other neuronal properties. Thus, constructing a comprehensive connectivity map requires associating various properties of neurons, including their connectivity, at cellular resolution. A commonly used approach is to use the gene expression profiles as an anchor to which all other neuronal properties are associated. Recent advances in genomics and anatomical techniques dramatically improved the ability to determine and associate the long-range projections of neurons with their gene expression profiles. These studies revealed unprecedented details of the gene-projection relationship, but also highlighted conceptual challenges in understanding this relationship. In this article, I delve into the findings and the challenges revealed by recent studies using state-of-the-art neuroanatomical and transcriptomic techniques. Building upon these insights, I propose an approach that focuses on understanding the gene-projection relationship through basic features in gene expression profiles and projections, respectively, that associate with underlying cellular processes. I then discuss how the developmental trajectories of projections and gene expression profiles create additional challenges and necessitate interrogating the gene-projection relationship across time. Finally, I explore complementary strategies that, together, can provide a comprehensive view of the gene-projection relationship.
Collapse
Affiliation(s)
- Xiaoyin Chen
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| |
Collapse
|
27
|
Barron JJ, Mroz NM, Taloma SE, Dahlgren MW, Ortiz-Carpena JF, Keefe MG, Escoubas CC, Dorman LC, Vainchtein ID, Chiaranunt P, Kotas ME, Nowakowski TJ, Bender KJ, Molofsky AB, Molofsky AV. Group 2 innate lymphoid cells promote inhibitory synapse development and social behavior. Science 2024; 386:eadi1025. [PMID: 39480923 PMCID: PMC11995778 DOI: 10.1126/science.adi1025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 02/22/2024] [Accepted: 09/02/2024] [Indexed: 11/02/2024]
Abstract
The innate immune system shapes brain development and is implicated in neurodevelopmental diseases. It is critical to define the relevant immune cells and signals and their impact on brain circuits. In this work, we found that group 2 innate lymphoid cells (ILC2s) and their cytokine interleukin-13 (IL-13) signaled directly to inhibitory interneurons to increase inhibitory synapse density in the developing mouse brain. ILC2s expanded and produced IL-13 in the developing brain meninges. Loss of ILC2s or IL-13 signaling to interneurons decreased inhibitory, but not excitatory, cortical synapses. Conversely, ILC2s and IL-13 were sufficient to increase inhibitory synapses. Loss of this signaling pathway led to selective impairments in social interaction. These data define a type 2 neuroimmune circuit in early life that shapes inhibitory synapse development and behavior.
Collapse
Affiliation(s)
- Jerika J. Barron
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nicholas M. Mroz
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sunrae E. Taloma
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
- Kavli Institute for Fundamental Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Madelene W. Dahlgren
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Lung biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Jorge F. Ortiz-Carpena
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Matthew G. Keefe
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Caroline C. Escoubas
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Leah C. Dorman
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Ilia D. Vainchtein
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Pailin Chiaranunt
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Maya E. Kotas
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tomasz J. Nowakowski
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kevin J. Bender
- Kavli Institute for Fundamental Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ari B. Molofsky
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Anna V. Molofsky
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Kavli Institute for Fundamental Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
28
|
Hong I, Kim J, Hainmueller T, Kim DW, Keijser J, Johnson RC, Park SH, Limjunyawong N, Yang Z, Cheon D, Hwang T, Agarwal A, Cholvin T, Krienen FM, McCarroll SA, Dong X, Leopold DA, Blackshaw S, Sprekeler H, Bergles DE, Bartos M, Brown SP, Huganir RL. Calcium-permeable AMPA receptors govern PV neuron feature selectivity. Nature 2024; 635:398-405. [PMID: 39358515 PMCID: PMC11560848 DOI: 10.1038/s41586-024-08027-2] [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: 07/19/2023] [Accepted: 09/05/2024] [Indexed: 10/04/2024]
Abstract
The brain helps us survive by forming internal representations of the external world1,2. Excitatory cortical neurons are often precisely tuned to specific external stimuli3,4. However, inhibitory neurons, such as parvalbumin-positive (PV) interneurons, are generally less selective5. PV interneurons differ from excitatory neurons in their neurotransmitter receptor subtypes, including AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) receptors (AMPARs)6,7. Excitatory neurons express calcium-impermeable AMPARs that contain the GluA2 subunit (encoded by GRIA2), whereas PV interneurons express receptors that lack the GluA2 subunit and are calcium-permeable (CP-AMPARs). Here we demonstrate a causal relationship between CP-AMPAR expression and the low feature selectivity of PV interneurons. We find low expression stoichiometry of GRIA2 mRNA relative to other subunits in PV interneurons that is conserved across ferrets, rodents, marmosets and humans, and causes abundant CP-AMPAR expression. Replacing CP-AMPARs in PV interneurons with calcium-impermeable AMPARs increased their orientation selectivity in the visual cortex. Manipulations to induce sparse CP-AMPAR expression demonstrated that this increase was cell-autonomous and could occur with changes beyond development. Notably, excitatory-PV interneuron connectivity rates and unitary synaptic strength were unaltered by CP-AMPAR removal, which suggested that the selectivity of PV interneurons can be altered without markedly changing connectivity. In Gria2-knockout mice, in which all AMPARs are calcium-permeable, excitatory neurons showed significantly degraded orientation selectivity, which suggested that CP-AMPARs are sufficient to drive lower selectivity regardless of cell type. Moreover, hippocampal PV interneurons, which usually exhibit low spatial tuning, became more spatially selective after removing CP-AMPARs, which indicated that CP-AMPARs suppress the feature selectivity of PV interneurons independent of modality. These results reveal a new role of CP-AMPARs in maintaining low-selectivity sensory representation in PV interneurons and implicate a conserved molecular mechanism that distinguishes this cell type in the neocortex.
Collapse
Affiliation(s)
- Ingie Hong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA.
| | - Juhyun Kim
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Thomas Hainmueller
- Institute for Physiology I, University of Freiburg, Medical Faculty, Freiburg, Germany
- Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Dong Won Kim
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Joram Keijser
- Modelling of Cognitive Processes, Technical University of Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Richard C Johnson
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Soo Hyun Park
- Section on Cognitive Neurophysiology and Imaging, National Institute of Mental Health, Bethesda, MD, USA
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Nathachit Limjunyawong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center of Research Excellence in Allergy and Immunology, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Zhuonan Yang
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - David Cheon
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Taeyoung Hwang
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amit Agarwal
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg, Germany
- Interdisciplinary Center for Neurosciences, University of Heidelberg, Heidelberg, Germany
| | - Thibault Cholvin
- Institute for Physiology I, University of Freiburg, Medical Faculty, Freiburg, Germany
| | - Fenna M Krienen
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | | | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David A Leopold
- Section on Cognitive Neurophysiology and Imaging, National Institute of Mental Health, Bethesda, MD, USA
| | - Seth Blackshaw
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Henning Sprekeler
- Modelling of Cognitive Processes, Technical University of Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Science of Intelligence, Research Cluster of Excellence, Berlin, Germany
| | - Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Marlene Bartos
- Institute for Physiology I, University of Freiburg, Medical Faculty, Freiburg, Germany
| | - Solange P Brown
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Richard L Huganir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
29
|
Keijser J, Hertäg L, Sprekeler H. Transcriptomic Correlates of State Modulation in GABAergic Interneurons: A Cross-Species Analysis. J Neurosci 2024; 44:e2371232024. [PMID: 39299800 PMCID: PMC11529809 DOI: 10.1523/jneurosci.2371-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/06/2024] [Accepted: 08/13/2024] [Indexed: 09/22/2024] Open
Abstract
GABAergic inhibitory interneurons comprise many subtypes that differ in their molecular, anatomical, and functional properties. In mouse visual cortex, they also differ in their modulation with an animal's behavioral state, and this state modulation can be predicted from the first principal component (PC) of the gene expression matrix. Here, we ask whether this link between transcriptome and state-dependent processing generalizes across species. To this end, we analysed seven single-cell and single-nucleus RNA sequencing datasets from mouse, human, songbird, and turtle forebrains. Despite homology at the level of cell types, we found clear differences between transcriptomic PCs, with greater dissimilarities between evolutionarily distant species. These dissimilarities arise from two factors: divergence in gene expression within homologous cell types and divergence in cell-type abundance. We also compare the expression of cholinergic receptors, which are thought to causally link transcriptome and state modulation. Several cholinergic receptors predictive of state modulation in mouse interneurons are differentially expressed between species. Circuit modelling and mathematical analyses suggest conditions under which these expression differences could translate into functional differences.
Collapse
Affiliation(s)
- Joram Keijser
- Modelling of Cognitive Processes, Technical University of Berlin, 10587 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, 10117 Berlin, Germany
| | - Loreen Hertäg
- Modelling of Cognitive Processes, Technical University of Berlin, 10587 Berlin, Germany
| | - Henning Sprekeler
- Modelling of Cognitive Processes, Technical University of Berlin, 10587 Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, 10115 Berlin, Germany
| |
Collapse
|
30
|
Arroyo B, Hernandez-Lemus E, Gutierrez R. The flow of reward information through neuronal ensembles in the accumbens. Cell Rep 2024; 43:114838. [PMID: 39395166 DOI: 10.1016/j.celrep.2024.114838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/05/2024] [Accepted: 09/20/2024] [Indexed: 10/14/2024] Open
Abstract
The nucleus accumbens shell (NAcSh) integrates reward information through diverse and specialized neuronal ensembles, influencing decision-making. By training rats in a probabilistic choice task and recording NAcSh neuronal activity, we found that rats adapt their choices based solely on the presence or absence of a sucrose reward, suggesting they build an internal representation of reward likelihood. We further demonstrate that NAcSh ensembles dynamically process different aspects of reward-guided behavior, with changes in composition and functional connections observed throughout the reinforcement learning process. The NAcSh forms a highly connected network characterized by a heavy-tailed distribution and the presence of neuronal hubs, facilitating efficient information flow. Reward delivery enhances mutual information, indicating increased communication between ensembles and network synchronization, whereas reward omission decreases it. Our findings reveal how reward information flows through dynamic NAcSh ensembles, whose flexible membership adapts as the rat learns to obtain rewards (energy) in an ever-changing environment.
Collapse
Affiliation(s)
- Benjamin Arroyo
- Laboratory Neurobiology of Appetite, Department of Pharmacology, CINVESTAV, Mexico City 07360, Mexico; Laboratory Neurobiology of Appetite, Center for Research on Aging (CIE), Cinvestav Sede Sur, Mexico City 14330, Mexico
| | - Enrique Hernandez-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; Center for Complexity Sciences, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
| | - Ranier Gutierrez
- Laboratory Neurobiology of Appetite, Department of Pharmacology, CINVESTAV, Mexico City 07360, Mexico; Laboratory Neurobiology of Appetite, Center for Research on Aging (CIE), Cinvestav Sede Sur, Mexico City 14330, Mexico.
| |
Collapse
|
31
|
Gao Y, van Velthoven CTJ, Lee C, Thomas ED, Bertagnolli D, Carey D, Casper T, Chakka AB, Chakrabarty R, Clark M, Desierto MJ, Ferrer R, Gloe J, Goldy J, Guilford N, Guzman J, Halterman CR, Hirschstein D, Ho W, James K, McCue R, Meyerdierks E, Nguy B, Pena N, Pham T, Shapovalova NV, Sulc J, Torkelson A, Tran A, Tung H, Wang J, Ronellenfitch K, Levi B, Hawrylycz MJ, Pagan C, Dee N, Smith KA, Tasic B, Yao Z, Zeng H. Continuous cell type diversification throughout the embryonic and postnatal mouse visual cortex development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.02.616246. [PMID: 39829740 PMCID: PMC11741437 DOI: 10.1101/2024.10.02.616246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The mammalian cortex is composed of a highly diverse set of cell types and develops through a series of temporally regulated events that build out the cell type and circuit foundation for cortical function. The mechanisms underlying the development of different cell types remain elusive. Single-cell transcriptomics provides the capacity to systematically study cell types across the entire temporal range of cortical development. Here, we present a comprehensive and high-resolution transcriptomic and epigenomic cell type atlas of the developing mouse visual cortex. The atlas was built from a single-cell RNA-sequencing dataset of 568,674 high-quality single-cell transcriptomes and a single-nucleus Multiome dataset of 194,545 high-quality nuclei providing both transcriptomic and chromatin accessibility profiles, densely sampled throughout the embryonic and postnatal developmental stages from E11.5 to P56. We computationally reconstructed a transcriptomic developmental trajectory map of all excitatory, inhibitory, and non-neuronal cell types in the visual cortex, identifying branching points marking the emergence of new cell types at specific developmental ages and defining molecular signatures of cellular diversification. In addition to neurogenesis, gliogenesis and early postmitotic maturation in the embryonic stage which gives rise to all the cell classes and nearly all subclasses, we find that increasingly refined cell types emerge throughout the postnatal differentiation process, including the late emergence of many cell types during the eye-opening stage (P11-P14) and the onset of critical period (P21), suggesting continuous cell type diversification at different stages of cortical development. Throughout development, we find cooperative dynamic changes in gene expression and chromatin accessibility in specific cell types, identifying both chromatin peaks potentially regulating the expression of specific genes and transcription factors potentially regulating specific peaks. Furthermore, a single gene can be regulated by multiple peaks associated with different cell types and/or different developmental stages. Collectively, our study provides the most detailed dynamic molecular map directly associated with individual cell types and specific developmental events that reveals the molecular logic underlying the continuous refinement of cell type identities in the developing visual cortex.
Collapse
Affiliation(s)
- Yuan Gao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Daniel Carey
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Rachel McCue
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Beagan Nguy
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Pena
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Alex Tran
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Justin Wang
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Boaz Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
32
|
Potter CT, Bassi CD, Runyan CA. Simultaneous interneuron labeling reveals cell type-specific, population-level interactions in cortex. iScience 2024; 27:110736. [PMID: 39280622 PMCID: PMC11399611 DOI: 10.1016/j.isci.2024.110736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/28/2024] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Cortical interneurons shape network activity in cell type-specific ways, and interact with other cell types. These interactions are understudied, as current methods typically restrict in vivo labeling to one neuron type. Although post-hoc identification of many cell types has been accomplished, the method is not available to many labs. We present a method to distinguish two red fluorophores in vivo, allowing imaging of activity in somatostatin (SOM), parvalbumin (PV), and the rest of the neural population in mouse cortex. We compared population events in PV and SOM neurons and observed that local network states reflected the ratio of SOM to PV neuron activity, demonstrating the importance of simultaneous labeling to explain dynamics. Activity became sparser and less correlated when the ratio between SOM and PV activity was high. Our simple method can be flexibly applied to study interactions among any combination of distinct cell type populations across brain areas.
Collapse
Affiliation(s)
- Christian T. Potter
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Constanza D. Bassi
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Caroline A. Runyan
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
33
|
Shi J, Nutkovich B, Kushinsky D, Rao BY, Herrlinger SA, Tsivourakis E, Mihaila TS, Paredes MEC, Malina KCK, O’Toole CK, Yong HC, Sanner BM, Xie A, Varol E, Losonczy A, Spiegel I. 2P-NucTag: on-demand phototagging for molecular analysis of functionally identified cortical neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586118. [PMID: 38585980 PMCID: PMC10996538 DOI: 10.1101/2024.03.21.586118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Neural circuits are characterized by genetically and functionally diverse cell types. A mechanistic understanding of circuit function is predicated on linking the genetic and physiological properties of individual neurons. However, it remains highly challenging to map the transcriptional properties to functionally heterogeneous neuronal subtypes in mammalian cortical circuits in vivo. Here, we introduce a high-throughput two-photon nuclear phototagging (2P-NucTag) approach optimized for on-demand and indelible labeling of single neurons via a photoactivatable red fluorescent protein following in vivo functional characterization in behaving mice. We demonstrate the utility of this function-forward pipeline by selectively labeling and transcriptionally profiling previously inaccessible 'place' and 'silent' cells in the mouse hippocampus. Our results reveal unexpected differences in gene expression between these hippocampal pyramidal neurons with distinct spatial coding properties. Thus, 2P-NucTag opens a new way to uncover the molecular principles that govern the functional organization of neural circuits.
Collapse
Affiliation(s)
- Jingcheng Shi
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, NY, United States
| | - Boaz Nutkovich
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Dahlia Kushinsky
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Bovey Y. Rao
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, NY, United States
| | - Stephanie A. Herrlinger
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Emmanouil Tsivourakis
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Tiberiu S. Mihaila
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Margaret E. Conde Paredes
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, NY, United States
- Tandon School of Engineering, New York University, New York, NY, United States
| | - Katayun Cohen-Kashi Malina
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Cliodhna K. O’Toole
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Hyun Choong Yong
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Brynn M. Sanner
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Angel Xie
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Erdem Varol
- Tandon School of Engineering, New York University, New York, NY, United States
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Ivo Spiegel
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
34
|
Lewis CM, Hoffmann A, Helmchen F. Linking brain activity across scales with simultaneous opto- and electrophysiology. NEUROPHOTONICS 2024; 11:033403. [PMID: 37662552 PMCID: PMC10472193 DOI: 10.1117/1.nph.11.3.033403] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023]
Abstract
The brain enables adaptive behavior via the dynamic coordination of diverse neuronal signals across spatial and temporal scales: from fast action potential patterns in microcircuits to slower patterns of distributed activity in brain-wide networks. Understanding principles of multiscale dynamics requires simultaneous monitoring of signals in multiple, distributed network nodes. Combining optical and electrical recordings of brain activity is promising for collecting data across multiple scales and can reveal aspects of coordinated dynamics invisible to standard, single-modality approaches. We review recent progress in combining opto- and electrophysiology, focusing on mouse studies that shed new light on the function of single neurons by embedding their activity in the context of brain-wide activity patterns. Optical and electrical readouts can be tailored to desired scales to tackle specific questions. For example, fast dynamics in single cells or local populations recorded with multi-electrode arrays can be related to simultaneously acquired optical signals that report activity in specified subpopulations of neurons, in non-neuronal cells, or in neuromodulatory pathways. Conversely, two-photon imaging can be used to densely monitor activity in local circuits while sampling electrical activity in distant brain areas at the same time. The refinement of combined approaches will continue to reveal previously inaccessible and under-appreciated aspects of coordinated brain activity.
Collapse
Affiliation(s)
| | - Adrian Hoffmann
- University of Zurich, Brain Research Institute, Zurich, Switzerland
- University of Zurich, Neuroscience Center Zurich, Zurich, Switzerland
| | - Fritjof Helmchen
- University of Zurich, Brain Research Institute, Zurich, Switzerland
- University of Zurich, Neuroscience Center Zurich, Zurich, Switzerland
- University of Zurich, University Research Priority Program, Adaptive Brain Circuits in Development and Learning, Zurich, Switzerland
| |
Collapse
|
35
|
Marghi Y, Gala R, Baftizadeh F, Sümbül U. Joint inference of discrete cell types and continuous type-specific variability in single-cell datasets with MMIDAS. NATURE COMPUTATIONAL SCIENCE 2024; 4:706-722. [PMID: 39317764 DOI: 10.1038/s43588-024-00683-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 08/06/2024] [Indexed: 09/26/2024]
Abstract
Reproducible definition and identification of cell types is essential to enable investigations into their biological function and to understand their relevance in the context of development, disease and evolution. Current approaches model variability in data as continuous latent factors, followed by clustering as a separate step, or immediately apply clustering on the data. We show that such approaches can suffer from qualitative mistakes in identifying cell types robustly, particularly when the number of such cell types is in the hundreds or even thousands. Here we propose an unsupervised method, Mixture Model Inference with Discrete-coupled AutoencoderS (MMIDAS), which combines a generalized mixture model with a multi-armed deep neural network to jointly infer the discrete type and continuous type-specific variability. Using four recent datasets of brain cells spanning different technologies, species and conditions, we demonstrate that MMIDAS can identify reproducible cell types and infer cell type-dependent continuous variability in both unimodal and multimodal datasets.
Collapse
Affiliation(s)
| | | | | | - Uygar Sümbül
- Allen Institute, Seattle, WA, USA.
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
36
|
Abbaspoor S, Hoffman KL. Circuit dynamics of superficial and deep CA1 pyramidal cells and inhibitory cells in freely moving macaques. Cell Rep 2024; 43:114519. [PMID: 39018243 PMCID: PMC11445748 DOI: 10.1016/j.celrep.2024.114519] [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: 01/21/2024] [Revised: 05/23/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024] Open
Abstract
Diverse neuron classes in hippocampal CA1 have been identified through the heterogeneity of their cellular/molecular composition. How these classes relate to hippocampal function and the network dynamics that support cognition in primates remains unclear. Here, we report inhibitory functional cell groups in CA1 of freely moving macaques whose diverse response profiles to network states and each other suggest distinct and specific roles in the functional microcircuit of CA1. In addition, pyramidal cells that were grouped by their superficial or deep layer position differed in firing rate, burstiness, and sharp-wave ripple-associated firing. They also showed strata-specific spike-timing interactions with inhibitory cell groups, suggestive of segregated neural populations. Furthermore, ensemble recordings revealed that cell assemblies were preferentially organized according to these strata. These results suggest that hippocampal CA1 in freely moving macaques bears a sublayer-specific circuit organization that may shape its role in cognition.
Collapse
Affiliation(s)
- Saman Abbaspoor
- Department of Psychology, Vanderbilt Vision Research Center, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
| | - Kari L Hoffman
- Department of Psychology, Vanderbilt Vision Research Center, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
37
|
Lim H, Zhang Y, Peters C, Straub T, Mayer JL, Klein R. Genetically- and spatially-defined basolateral amygdala neurons control food consumption and social interaction. Nat Commun 2024; 15:6868. [PMID: 39127719 PMCID: PMC11316773 DOI: 10.1038/s41467-024-50889-7] [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: 01/04/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
The basolateral amygdala (BLA) contains discrete neuronal circuits that integrate positive or negative emotional information and drive the appropriate innate and learned behaviors. Whether these circuits consist of genetically-identifiable and anatomically segregated neuron types, is poorly understood. Also, our understanding of the response patterns and behavioral spectra of genetically-identifiable BLA neurons is limited. Here, we classified 11 glutamatergic cell clusters in mouse BLA and found that several of them were anatomically segregated in lateral versus basal amygdala, and anterior versus posterior regions of the BLA. Two of these BLA subpopulations innately responded to valence-specific, whereas one responded to mixed - aversive and social - cues. Positive-valence BLA neurons promoted normal feeding, while mixed selectivity neurons promoted fear learning and social interactions. These findings enhance our understanding of cell type diversity and spatial organization of the BLA and the role of distinct BLA populations in representing valence-specific and mixed stimuli.
Collapse
Affiliation(s)
- Hansol Lim
- Department Molecules - Signaling - Development, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Yue Zhang
- Department Synapses - Circuits - Plasticity, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Christian Peters
- Department Molecules - Signaling - Development, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Tobias Straub
- Biomedical Center Core Facility Bioinformatics, LMU, Munich, Germany
| | - Johanna Luise Mayer
- Department Molecules - Signaling - Development, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Rüdiger Klein
- Department Molecules - Signaling - Development, Max Planck Institute for Biological Intelligence, Martinsried, Germany.
| |
Collapse
|
38
|
Machold R, Rudy B. Genetic approaches to elucidating cortical and hippocampal GABAergic interneuron diversity. Front Cell Neurosci 2024; 18:1414955. [PMID: 39113758 PMCID: PMC11303334 DOI: 10.3389/fncel.2024.1414955] [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: 04/09/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
GABAergic interneurons (INs) in the mammalian forebrain represent a diverse population of cells that provide specialized forms of local inhibition to regulate neural circuit activity. Over the last few decades, the development of a palette of genetic tools along with the generation of single-cell transcriptomic data has begun to reveal the molecular basis of IN diversity, thereby providing deep insights into how different IN subtypes function in the forebrain. In this review, we outline the emerging picture of cortical and hippocampal IN speciation as defined by transcriptomics and developmental origin and summarize the genetic strategies that have been utilized to target specific IN subtypes, along with the technical considerations inherent to each approach. Collectively, these methods have greatly facilitated our understanding of how IN subtypes regulate forebrain circuitry via cell type and compartment-specific inhibition and thus have illuminated a path toward potential therapeutic interventions for a variety of neurocognitive disorders.
Collapse
Affiliation(s)
- Robert Machold
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Bernardo Rudy
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
39
|
Agmon A, Barth AL. A brief history of somatostatin interneuron taxonomy or: how many somatostatin subtypes are there, really? Front Neural Circuits 2024; 18:1436915. [PMID: 39091993 PMCID: PMC11292610 DOI: 10.3389/fncir.2024.1436915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 06/28/2024] [Indexed: 08/04/2024] Open
Abstract
We provide a brief (and unabashedly biased) overview of the pre-transcriptomic history of somatostatin interneuron taxonomy, followed by a chronological summary of the large-scale, NIH-supported effort over the last ten years to generate a comprehensive, single-cell RNA-seq-based taxonomy of cortical neurons. Focusing on somatostatin interneurons, we present the perspective of experimental neuroscientists trying to incorporate the new classification schemes into their own research while struggling to keep up with the ever-increasing number of proposed cell types, which seems to double every two years. We suggest that for experimental analysis, the most useful taxonomic level is the subdivision of somatostatin interneurons into ten or so "supertypes," which closely agrees with their more traditional classification by morphological, electrophysiological and neurochemical features. We argue that finer subdivisions ("t-types" or "clusters"), based on slight variations in gene expression profiles but lacking clear phenotypic differences, are less useful to researchers and may actually defeat the purpose of classifying neurons to begin with. We end by stressing the need for generating novel tools (mouse lines, viral vectors) for genetically targeting distinct supertypes for expression of fluorescent reporters, calcium sensors and excitatory or inhibitory opsins, allowing neuroscientists to chart the input and output synaptic connections of each proposed subtype, reveal the position they occupy in the cortical network and examine experimentally their roles in sensorimotor behaviors and cognitive brain functions.
Collapse
Affiliation(s)
- Ariel Agmon
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Alison L. Barth
- Department of Biological Sciences, Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA, United States
| |
Collapse
|
40
|
Chemerkouh MJHN, Zhou X, Yang Y, Wang S. Deep Learning Enhanced Label-Free Action Potential Detection Using Plasmonic-Based Electrochemical Impedance Microscopy. Anal Chem 2024; 96:11299-11308. [PMID: 38953225 PMCID: PMC11283340 DOI: 10.1021/acs.analchem.4c01179] [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] [Indexed: 07/03/2024]
Abstract
Measuring neuronal electrical activity, such as action potential propagation in cells, requires the sensitive detection of the weak electrical signal with high spatial and temporal resolution. None of the existing tools can fulfill this need. Recently, plasmonic-based electrochemical impedance microscopy (P-EIM) was demonstrated for the label-free mapping of the ignition and propagation of action potentials in neuron cells with subcellular resolution. However, limited by the signal-to-noise ratio in the high-speed P-EIM video, action potential mapping was achieved by averaging 90 cycles of signals. Such extensive averaging is not desired and may not always be feasible due to factors such as neuronal desensitization. In this study, we utilized advanced signal processing techniques to detect action potentials in P-EIM extracted signals with fewer averaged cycles. Matched filtering successfully detected action potential signals with as few as averaging five cycles of signals. Long short-term memory (LSTM) recurrent neural network achieved the best performance and was able to detect single-cycle stimulated action potential successfully [satisfactory area under the receiver operating characteristic curve (AUC) equal to 0.855]. Therefore, we show that deep learning-based signal processing can dramatically improve the usability of P-EIM mapping of neuronal electrical signals.
Collapse
Affiliation(s)
- Mohammad Javad Haji Najafi Chemerkouh
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ 85287, USA
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Xinyu Zhou
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ 85287, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Yunze Yang
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ 85287, USA
| | - Shaopeng Wang
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ 85287, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
41
|
Huang S, Rizzo D, Wu SJ, Xu Q, Ziane L, Alghamdi N, Stafford DA, Daigle TL, Tasic B, Zeng H, Ibrahim LA, Fishell G. Neurogliaform Cells Exhibit Laminar-specific Responses in the Visual Cortex and Modulate Behavioral State-dependent Cortical Activity. RESEARCH SQUARE 2024:rs.3.rs-4530873. [PMID: 39011116 PMCID: PMC11247929 DOI: 10.21203/rs.3.rs-4530873/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Neurogliaform cells are a distinct type of GABAergic cortical interneurons known for their 'volume transmission' output property. However, their activity and function within cortical circuits remain unclear. Here, we developed two genetic tools to target these neurons and examine their function in the primary visual cortex. We found that the spontaneous activity of neurogliaform cells positively correlated with locomotion. Silencing these neurons increased spontaneous activity during locomotion and impaired visual responses in L2/3 pyramidal neurons. Furthermore, the contrast-dependent visual response of neurogliaform cells varies with their laminar location and is constrained by their morphology and input connectivity. These findings demonstrate the importance of neurogliaform cells in regulating cortical behavioral state-dependent spontaneous activity and indicate that their functional engagement during visual stimuli is influenced by their laminar positioning and connectivity.
Collapse
Affiliation(s)
- Shuhan Huang
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Program in Neuroscience, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniella Rizzo
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sherry Jingjing Wu
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Qing Xu
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Past address: Center for Genomics & Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Leena Ziane
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Norah Alghamdi
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - David A Stafford
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94708, USA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Leena Ali Ibrahim
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Gord Fishell
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
42
|
Marghi Y, Gala R, Baftizadeh F, Sümbül U. Joint inference of discrete cell types and continuous type-specific variability in single-cell datasets with MMIDAS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.02.560574. [PMID: 37873271 PMCID: PMC10592946 DOI: 10.1101/2023.10.02.560574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Reproducible definition and identification of cell types is essential to enable investigations into their biological function, and understanding their relevance in the context of development, disease and evolution. Current approaches model variability in data as continuous latent factors, followed by clustering as a separate step, or immediately apply clustering on the data. We show that such approaches can suffer from qualitative mistakes in identifying cell types robustly, particularly when the number of such cell types is in the hundreds or even thousands. Here, we propose an unsupervised method, MMIDAS, which combines a generalized mixture model with a multi-armed deep neural network, to jointly infer the discrete type and continuous type-specific variability. Using four recent datasets of brain cells spanning different technologies, species, and conditions, we demonstrate that MMIDAS can identify reproducible cell types and infer cell type-dependent continuous variability in both uni-modal and multi-modal datasets.
Collapse
Affiliation(s)
| | - Rohan Gala
- Allen Institute, 615 Westlake Ave N, Seattle, WA, USA
| | | | - Uygar Sümbül
- Allen Institute, 615 Westlake Ave N, Seattle, WA, USA
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
43
|
Ostojic S, Fusi S. Computational role of structure in neural activity and connectivity. Trends Cogn Sci 2024; 28:677-690. [PMID: 38553340 DOI: 10.1016/j.tics.2024.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 07/05/2024]
Abstract
One major challenge of neuroscience is identifying structure in seemingly disorganized neural activity. Different types of structure have different computational implications that can help neuroscientists understand the functional role of a particular brain area. Here, we outline a unified approach to characterize structure by inspecting the representational geometry and the modularity properties of the recorded activity and show that a similar approach can also reveal structure in connectivity. We start by setting up a general framework for determining geometry and modularity in activity and connectivity and relating these properties with computations performed by the network. We then use this framework to review the types of structure found in recent studies of model networks performing three classes of computations.
Collapse
Affiliation(s)
- Srdjan Ostojic
- Laboratoire de Neurosciences Cognitives et Computationnelles, INSERM U960, Ecole Normale Superieure - PSL Research University, 75005 Paris, France.
| | - Stefano Fusi
- Center for Theoretical Neuroscience, Columbia University, New York, NY, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Neuroscience, Columbia University, New York, NY, USA; Kavli Institute for Brain Science, Columbia University, New York, NY, USA
| |
Collapse
|
44
|
Ratliff JM, Terral G, Lutzu S, Heiss J, Mota J, Stith B, Lechuga AV, Ramakrishnan C, Fenno LE, Daigle T, Deisseroth K, Zeng H, Ngai J, Tasic B, Sjulson L, Rudolph S, Kilduff TS, Batista-Brito R. Neocortical long-range inhibition promotes cortical synchrony and sleep. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599756. [PMID: 38948753 PMCID: PMC11213009 DOI: 10.1101/2024.06.20.599756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Behavioral states such as sleep and wake are highly correlated with specific patterns of rhythmic activity in the cortex. During low arousal states such as slow wave sleep, the cortex is synchronized and dominated by low frequency rhythms coordinated across multiple regions. Although recent evidence suggests that GABAergic inhibitory neurons are key players in cortical state modulation, the in vivo circuit mechanisms coordinating synchronized activity among local and distant neocortical networks are not well understood. Here, we show that somatostatin and chondrolectin co-expressing cells (Sst-Chodl cells), a sparse and unique class of neocortical inhibitory neurons, are selectively active during low arousal states and are largely silent during periods of high arousal. In contrast to other neocortical inhibitory neurons, we show these neurons have long-range axons that project across neocortical areas. Activation of Sst-Chodl cells is sufficient to promote synchronized cortical states characteristic of low arousal, with increased spike co-firing and low frequency brain rhythms, and to alter behavioral states by promoting sleep. Contrary to the prevailing belief that sleep is exclusively driven by subcortical mechanisms, our findings reveal that these long-range inhibitory neurons not only track changes in behavioral state but are sufficient to induce both sleep-like cortical states and sleep behavior, establishing a crucial circuit component in regulating behavioral states.
Collapse
Affiliation(s)
- Jacob M Ratliff
- Albert Einstein College of Medicine, New York City, NY, United States
| | - Geoffrey Terral
- Albert Einstein College of Medicine, New York City, NY, United States
| | - Stefano Lutzu
- Albert Einstein College of Medicine, New York City, NY, United States
| | - Jaime Heiss
- Biosciences Division, SRI International, Menlo Park, CA 94025, United States
| | - Julie Mota
- Albert Einstein College of Medicine, New York City, NY, United States
| | - Bianca Stith
- Albert Einstein College of Medicine, New York City, NY, United States
| | | | | | - Lief E Fenno
- The University of Texas at Austin, Austin, TX, United States
| | - Tanya Daigle
- Allen Institute for Brain Science, Seattle, WA, United States
| | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, United States
| | - John Ngai
- National Institute of Neurological Disease and Stroke, Bethesda, MD, United States
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA, United States
| | - Lucas Sjulson
- Albert Einstein College of Medicine, New York City, NY, United States
| | - Stephanie Rudolph
- Albert Einstein College of Medicine, New York City, NY, United States
| | - Thomas S. Kilduff
- Biosciences Division, SRI International, Menlo Park, CA 94025, United States
| | | |
Collapse
|
45
|
Yoneda T, Kameyama K, Gotou T, Terata K, Takagi M, Yoshimura Y, Sakimura K, Kano M, Hata Y. Layer specific regulation of critical period timing and maturation of mouse visual cortex by endocannabinoids. iScience 2024; 27:110145. [PMID: 38952682 PMCID: PMC11215304 DOI: 10.1016/j.isci.2024.110145] [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: 05/25/2023] [Revised: 04/15/2024] [Accepted: 05/27/2024] [Indexed: 07/03/2024] Open
Abstract
Plasticity during the critical period is important for the functional maturation of cortical neurons. While characteristics of plasticity are diverse among cortical layers, it is unknown whether critical period timing is controlled by a common or unique molecular mechanism among them. We here clarified layer-specific regulation of the critical period timing of ocular dominance plasticity in the primary visual cortex. Mice lacking the endocannabinoid synthesis enzyme diacylglycerol lipase-α exhibited precocious critical period timing, earlier maturation of inhibitory synaptic function in layers 2/3 and 4, and impaired development of the binocular matching of orientation selectivity exclusively in layer 2/3. Activation of cannabinoid receptor restored ocular dominance plasticity at the normal critical period in layer 2/3. Suppression of GABAA receptor rescued precocious ocular dominance plasticity in layer 4. Therefore, endocannabinoids regulate critical period timing and maturation of visual function partly through the development of inhibitory synaptic functions in a layer-dependent manner.
Collapse
Affiliation(s)
- Taisuke Yoneda
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
- Division of Visual Information Processing, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Okazaki 444-8585, Japan
| | - Katsuro Kameyama
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Takahiro Gotou
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Keiko Terata
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Masahiro Takagi
- Division of Visual Information Processing, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Yumiko Yoshimura
- Division of Visual Information Processing, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Okazaki 444-8585, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo 113-0033, Japan
- Advanced Comprehensive Research Organization (ACRO), Teikyo University, Tokyo 173-0003, Japan
| | - Yoshio Hata
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| |
Collapse
|
46
|
Huang S, Rizzo D, Wu SJ, Xu Q, Ziane L, Alghamdi N, Stafford DA, Daigle TL, Tasic B, Zeng H, Ibrahim LA, Fishell G. Neurogliaform Cells Exhibit Laminar-specific Responses in the Visual Cortex and Modulate Behavioral State-dependent Cortical Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597539. [PMID: 38895403 PMCID: PMC11185653 DOI: 10.1101/2024.06.05.597539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Neurogliaform cells are a distinct type of GABAergic cortical interneurons known for their "volume transmission" output property. However, their activity and function within cortical circuits remain unclear. Here, we developed two genetic tools to target these neurons and examine their function in the primary visual cortex. We found that the spontaneous activity of neurogliaform cells positively correlated with locomotion. Silencing these neurons increased spontaneous activity during locomotion and impaired visual responses in L2/3 pyramidal neurons. Furthermore, the contrast-dependent visual response of neurogliaform cells varies with their laminar location and is constrained by their morphology and input connectivity. These findings demonstrate the importance of neurogliaform cells in regulating cortical behavioral state-dependent spontaneous activity and indicate that their functional engagement during visual stimuli is influenced by their laminar positioning and connectivity.
Collapse
Affiliation(s)
- Shuhan Huang
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Program in Neuroscience, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniella Rizzo
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sherry Jingjing Wu
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Qing Xu
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Past address: Center for Genomics & Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Leena Ziane
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Norah Alghamdi
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - David A Stafford
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94708, USA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Leena Ali Ibrahim
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Gord Fishell
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
47
|
Lee AT, Chang EF, Paredes MF, Nowakowski TJ. Large-scale neurophysiology and single-cell profiling in human neuroscience. Nature 2024; 630:587-595. [PMID: 38898291 PMCID: PMC12049086 DOI: 10.1038/s41586-024-07405-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 04/09/2024] [Indexed: 06/21/2024]
Abstract
Advances in large-scale single-unit human neurophysiology, single-cell RNA sequencing, spatial transcriptomics and long-term ex vivo tissue culture of surgically resected human brain tissue have provided an unprecedented opportunity to study human neuroscience. In this Perspective, we describe the development of these paradigms, including Neuropixels and recent brain-cell atlas efforts, and discuss how their convergence will further investigations into the cellular underpinnings of network-level activity in the human brain. Specifically, we introduce a workflow in which functionally mapped samples of human brain tissue resected during awake brain surgery can be cultured ex vivo for multi-modal cellular and functional profiling. We then explore how advances in human neuroscience will affect clinical practice, and conclude by discussing societal and ethical implications to consider. Potential findings from the field of human neuroscience will be vast, ranging from insights into human neurodiversity and evolution to providing cell-type-specific access to study and manipulate diseased circuits in pathology. This Perspective aims to provide a unifying framework for the field of human neuroscience as we welcome an exciting era for understanding the functional cytoarchitecture of the human brain.
Collapse
Affiliation(s)
- Anthony T Lee
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Edward F Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mercedes F Paredes
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
48
|
de Brito Van Velze M, Dhanasobhon D, Martinez M, Morabito A, Berthaux E, Pinho CM, Zerlaut Y, Rebola N. Feedforward and disinhibitory circuits differentially control activity of cortical somatostatin interneurons during behavioral state transitions. Cell Rep 2024; 43:114197. [PMID: 38733587 DOI: 10.1016/j.celrep.2024.114197] [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: 10/05/2023] [Revised: 03/26/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
Interneurons (INs), specifically those in disinhibitory circuits like somatostatin (SST) and vasoactive intestinal peptide (VIP)-INs, are strongly modulated by the behavioral context. Yet, the mechanisms by which these INs are recruited during active states and whether their activity is consistent across sensory cortices remain unclear. We now report that in mice, locomotor activity strongly recruits SST-INs in the primary somatosensory (S1) but not the visual (V1) cortex. This diverse engagement of SST-INs cannot be explained by differences in VIP-IN function but is absent in the presence of visual input, suggesting the involvement of feedforward sensory pathways. Accordingly, inactivating the somatosensory thalamus, but not decreasing VIP-IN activity, significantly reduces the modulation of SST-INs by locomotion. Model simulations suggest that the differences in SST-INs across behavioral states can be explained by varying ratios of VIP- and thalamus-driven activity. By integrating feedforward activity with neuromodulation, SST-INs are anticipated to be crucial for adapting sensory processing to behavioral states.
Collapse
Affiliation(s)
- Marcel de Brito Van Velze
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Dhanasak Dhanasobhon
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Marie Martinez
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Annunziato Morabito
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Emmanuelle Berthaux
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Cibele Martins Pinho
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, INSERM, CNRS, 75013 Paris, France
| | - Yann Zerlaut
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, INSERM, CNRS, 75013 Paris, France.
| | - Nelson Rebola
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, INSERM, CNRS, 75013 Paris, France.
| |
Collapse
|
49
|
Lee EJ, Suh M, Choi H, Choi Y, Hwang DW, Bae S, Lee DS. Spatial transcriptomic brain imaging reveals the effects of immunomodulation therapy on specific regional brain cells in a mouse dementia model. BMC Genomics 2024; 25:516. [PMID: 38796425 PMCID: PMC11128132 DOI: 10.1186/s12864-024-10434-8] [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: 02/18/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
Increasing evidence of brain-immune crosstalk raises expectations for the efficacy of novel immunotherapies in Alzheimer's disease (AD), but the lack of methods to examine brain tissues makes it difficult to evaluate therapeutics. Here, we investigated the changes in spatial transcriptomic signatures and brain cell types using the 10x Genomics Visium platform in immune-modulated AD models after various treatments. To proceed with an analysis suitable for barcode-based spatial transcriptomics, we first organized a workflow for segmentation of neuroanatomical regions, establishment of appropriate gene combinations, and comprehensive review of altered brain cell signatures. Ultimately, we investigated spatial transcriptomic changes following administration of immunomodulators, NK cell supplements and an anti-CD4 antibody, which ameliorated behavior impairment, and designated brain cells and regions showing probable associations with behavior changes. We provided the customized analytic pipeline into an application named STquantool. Thus, we anticipate that our approach can help researchers interpret the real action of drug candidates by simultaneously investigating the dynamics of all transcripts for the development of novel AD therapeutics.
Collapse
Affiliation(s)
- Eun Ji Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Minseok Suh
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yoori Choi
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Cliniclal Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Do Won Hwang
- Research and Development Center, THERABEST Inc., Seocho-daero 40-gil, Seoul, 06657, Republic of Korea
| | - Sungwoo Bae
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Medical Science and Engineering, School of Convergence Science and Technology, POSTECH, Pohang, Republic of Korea.
| |
Collapse
|
50
|
Abbaspoor S, Hoffman KL. Circuit dynamics of superficial and deep CA1 pyramidal cells and inhibitory cells in freely-moving macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.06.570369. [PMID: 38106053 PMCID: PMC10723348 DOI: 10.1101/2023.12.06.570369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Diverse neuron classes in hippocampal CA1 have been identified through the heterogeneity of their cellular/molecular composition. How these classes relate to hippocampal function and the network dynamics that support cognition in primates remains unclear. Here we report inhibitory functional cell groups in CA1 of freely-moving macaques whose diverse response profiles to network states and each other suggest distinct and specific roles in the functional microcircuit of CA1. In addition, pyramidal cells that were segregated into superficial and deep layers differed in firing rate, burstiness, and sharp-wave ripple-associated firing. They also showed strata-specific spike-timing interactions with inhibitory cell groups, suggestive of segregated neural populations. Furthermore, ensemble recordings revealed that cell assemblies were preferentially organized according to these strata. These results suggest sublayer-specific circuit organization in hippocampal CA1 of the freely-moving macaques that may underlie its role in cognition.
Collapse
Affiliation(s)
- S Abbaspoor
- Department of Psychology, Vanderbilt Vision Research Center, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee
| | - K L Hoffman
- Department of Psychology, Vanderbilt Vision Research Center, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|