1
|
Zhang XH, Anderson KM, Dong HM, Chopra S, Dhamala E, Emani PS, Gerstein MB, Margulies DS, Holmes AJ. The cell-type underpinnings of the human functional cortical connectome. Nat Neurosci 2025; 28:150-160. [PMID: 39572742 DOI: 10.1038/s41593-024-01812-2] [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: 07/27/2023] [Accepted: 09/26/2024] [Indexed: 11/27/2024]
Abstract
The functional properties of the human brain arise, in part, from the vast assortment of cell types that pattern the cerebral cortex. The cortical sheet can be broadly divided into distinct networks, which are embedded into processing streams, or gradients, that extend from unimodal systems through higher-order association territories. Here using microarray data from the Allen Human Brain Atlas and single-nucleus RNA-sequencing data from multiple cortical territories, we demonstrate that cell-type distributions are spatially coupled to the functional organization of cortex, as estimated through functional magnetic resonance imaging. Differentially enriched cells follow the spatial topography of both functional gradients and associated large-scale networks. Distinct cellular fingerprints were evident across networks, and a classifier trained on postmortem cell-type distributions was able to predict the functional network allegiance of cortical tissue samples. These data indicate that the in vivo organization of the cortical sheet is reflected in the spatial variability of its cellular composition.
Collapse
Affiliation(s)
- Xi-Han Zhang
- Department of Psychology, Yale University, New Haven, CT, USA.
| | | | - Hao-Ming Dong
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Sidhant Chopra
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Elvisha Dhamala
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Prashant S Emani
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Mark B Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA
| | - Daniel S Margulies
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Cognitive Neuroanatomy Lab, Université Paris Cité, INCC UMR 8002, CNRS, Paris, France
| | - Avram J Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
2
|
Wierda K, Nyitrai H, Lejeune A, Vlaeminck I, Leysen E, Theys T, de Wit J, Vanderhaeghen P, Libé-Philippot B. Protocol to process fresh human cerebral cortex biopsies for patch-clamp recording and immunostaining. STAR Protoc 2024; 5:103313. [PMID: 39292560 PMCID: PMC11424940 DOI: 10.1016/j.xpro.2024.103313] [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: 05/23/2024] [Revised: 07/23/2024] [Accepted: 08/22/2024] [Indexed: 09/20/2024] Open
Abstract
Cerebral cortex biopsies enable the investigation of native developing and mature human brain tissue. Here, we present a protocol to process human cortical biopsies from the surgical theater to the laboratory. We describe steps for the preparation of viable acute slices for patch-clamp recording using dedicated chemical solutions for transport and sectioning. We then explain procedures for tissue fixation and post hoc immunostaining to correlate physiological properties to morphological features and protein detection. For complete details on the use and execution of this protocol, please refer to Libé-Philippot et al.1.
Collapse
Affiliation(s)
- Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium.
| | - Hajnalka Nyitrai
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Amélie Lejeune
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Ine Vlaeminck
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Elke Leysen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Tom Theys
- KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; Research Group Experimental Neurosurgery and Neuroanatomy, KUL, 3000 Leuven, Belgium
| | - Joris de Wit
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium.
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium.
| | - Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium.
| |
Collapse
|
3
|
Kanari L, Shi Y, Arnaudon A, Barros-Zulaica N, Benavides-Piccione R, Coggan JS, DeFelipe J, Hess K, Mansvelder HD, Mertens EJ, Meystre J, de Campos Perin R, Pezzoli M, Daniel RT, Stoop R, Segev I, Markram H, de Kock CP. Of mice and men: Dendritic architecture differentiates human from mice neuronal networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.11.557170. [PMID: 39763990 PMCID: PMC11702562 DOI: 10.1101/2023.09.11.557170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
The organizational principles that distinguish the human brain from other species have been a long-standing enigma in neuroscience. Focusing on the uniquely evolved human cortical layers 2 and 3, we computationally reconstruct the cortical architecture for mice and humans. We show that human pyramidal cells form highly complex networks, demonstrated by the increased number and simplex dimension compared to mice. This is surprising because human pyramidal cells are much sparser in the cortex. We show that the number and size of neurons fail to account for this increased network complexity, suggesting that another morphological property is a key determinant of network connectivity. Topological comparison of dendritic structure reveals much higher perisomatic (basal and oblique) branching density in human pyramidal cells. Using topological tools we quantitatively show that this neuronal structural property directly impacts network complexity, including the formation of a rich subnetwork structure. We conclude that greater dendritic complexity, a defining attribute of human L2 and 3 neurons, may provide the human cortex with enhanced computational capacity and cognitive flexibility.
Collapse
Affiliation(s)
- Lida Kanari
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Ying Shi
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Alexis Arnaudon
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Natalí Barros-Zulaica
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Ruth Benavides-Piccione
- Laboratorio Cajal de Circuitos Corticales, Universidad Politécnica de Madrid and Instituto Cajal (CSIC), Pozuelo de Alarcón, Madrid 28223, Spain
| | - Jay S. Coggan
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Universidad Politécnica de Madrid and Instituto Cajal (CSIC), Pozuelo de Alarcón, Madrid 28223, Spain
| | - Kathryn Hess
- Laboratory for Topology and Neuroscience, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Huib D. Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Eline J. Mertens
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Julie Meystre
- Laboratory of Neural Microcircuitry, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Rodrigo de Campos Perin
- Laboratory of Neural Microcircuitry, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Maurizio Pezzoli
- Laboratory of Neural Microcircuitry, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Roy Thomas Daniel
- Department of Clinical Neurosciences, Neurosurgery Unit, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Ron Stoop
- Center for Psychiatric Neurosciences, Department of Psychiatry, Lausanne University Hospital Center, Lausanne, Switzerland
| | - Idan Segev
- Department of Neurobiology and Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, 9190501 Jerusalem, Israel
| | - Henry Markram
- Blue Brain Project, École Polytechnique Fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Christiaan P.J. de Kock
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| |
Collapse
|
4
|
Libé-Philippot B, Polleux F, Vanderhaeghen P. If you please, draw me a neuron - linking evolutionary tinkering with human neuron evolution. Curr Opin Genet Dev 2024; 89:102260. [PMID: 39357501 PMCID: PMC11625661 DOI: 10.1016/j.gde.2024.102260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/23/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024]
Abstract
Animal speciation often involves novel behavioral features that rely on nervous system evolution. Human-specific brain features have been proposed to underlie specialized cognitive functions and to be linked, at least in part, to the evolution of synapses, neurons, and circuits of the cerebral cortex. Here, we review recent results showing that, while the human cortex is composed of a repertoire of cells that appears to be largely similar to the one found in other mammals, human cortical neurons do display specialized features at many levels, from gene expression to intrinsic physiological properties. The molecular mechanisms underlying human species-specific neuronal features remain largely unknown but implicate hominid-specific gene duplicates that encode novel molecular modifiers of neuronal function. The identification of human-specific genetic modifiers of neuronal function brings novel insights on brain evolution and function and, could also provide new insights on human species-specific vulnerabilities to brain disorders.
Collapse
Affiliation(s)
- Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium; Aix-Marseille Université, CNRS UMR 7288, Developmental Biology Institute of Marseille (IBDM), NeuroMarseille, Marseille, France.
| | - Franck Polleux
- Department of Neuroscience, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA. https://twitter.com/@fpolleux
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium.
| |
Collapse
|
5
|
Ting JT, Johansen NJ, Kalmbach BE, Taskin N, Lee B, Clark JK, Kendrick R, Ng L, Radaelli C, Weed N, Enstrom R, Ransford S, Redford I, Walling-Bell S, Dalley R, Tieu M, Goldy J, Jorstad N, Smith K, Bakken T, Lein ES, Owen SF. Distinctive physiology of molecularly identified medium spiny neurons in the macaque putamen. Cell Rep 2024; 43:114963. [PMID: 39514389 DOI: 10.1016/j.celrep.2024.114963] [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: 04/19/2024] [Revised: 09/11/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The distinctive physiology of striatal medium spiny neurons (MSNs) underlies their ability to integrate sensory and motor input. In rodents, MSNs have a hyperpolarized resting potential and low input resistance. When activated, they have a delayed onset of spiking and regular spike rate. Here, we show that in the macaque putamen, latency to spike is reduced and spike rate adaptation is increased relative to mouse. We use whole-cell brain slice recordings and recover single-cell gene expression using Patch-seq to distinguish macaque MSN cell types. Species differences in the expression of ion channel genes including the calcium-activated chloride channel, ANO2, and an auxiliary subunit of the A-type potassium channel, DPP10, are correlated with species differences in spike rate adaptation and latency to the first spike, respectively. These surprising divergences in physiology better define the strengths and limitations of mouse models for understanding neuronal and circuit function in the primate basal ganglia.
Collapse
Affiliation(s)
- Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA 98109, USA; The Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA; Department of Neurobiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | | | - Brian E Kalmbach
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Neurobiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brian Lee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jason K Clark
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rennie Kendrick
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lindsay Ng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rachel Enstrom
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Shea Ransford
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ingrid Redford
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Rachel Dalley
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Nik Jorstad
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kimberly Smith
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Trygve Bakken
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Scott F Owen
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
6
|
Caccavano AP, Vlachos A, McLean N, Kimmel S, Kim JH, Vargish G, Mahadevan V, Hewitt L, Rossi AM, Spineux I, Wu SJ, Furlanis E, Dai M, Garcia BL, Chittajallu R, London E, Yuan X, Hunt S, Abebe D, Eldridge MAG, Cummins AC, Hines BE, Plotnikova A, Mohanty A, Averbeck BB, Zaghloul K, Dimidschstein J, Fishell G, Pelkey KA, McBain CJ. Divergent opioid-mediated suppression of inhibition between hippocampus and neocortex across species and development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.20.576455. [PMID: 38313283 PMCID: PMC10836073 DOI: 10.1101/2024.01.20.576455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Opioid receptors within the CNS regulate pain sensation and mood and are key targets for drugs of abuse. Within the adult rodent hippocampus (HPC), μ-opioid receptor agonists suppress inhibitory parvalbumin-expressing interneurons (PV-INs), thus disinhibiting the circuit. However, it is uncertain if this disinhibitory motif is conserved in other cortical regions, species, or across development. We observed that PV-IN mediated inhibition is robustly suppressed by opioids in hippocampus proper but not neocortex in mice and nonhuman primates, with spontaneous inhibitory tone in resected human tissue also following a consistent dichotomy. This hippocampal disinhibitory motif was established in early development when PV-INs and opioids were found to regulate primordial network rhythmogenesis. Acute opioid-mediated modulation was partially occluded with morphine pretreatment, with implications for the effects of opioids on hippocampal network activity important for learning and memory. Together, these findings demonstrate that PV-INs exhibit a divergence in opioid sensitivity across brain regions that is remarkably conserved across evolution and highlights the underappreciated role of opioids acting through immature PV-INs in shaping hippocampal development.
Collapse
Affiliation(s)
- Adam P Caccavano
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anna Vlachos
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nadiya McLean
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sarah Kimmel
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - June Hoan Kim
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Geoffrey Vargish
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Vivek Mahadevan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Lauren Hewitt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anthony M Rossi
- 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
| | - Ilona Spineux
- 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
| | - Elisabetta Furlanis
- 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
| | - Min Dai
- 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
| | - Brenda Leyva Garcia
- 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
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Edra London
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaoqing Yuan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Steven Hunt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Daniel Abebe
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mark A G Eldridge
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Alex C Cummins
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Brendan E Hines
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Anya Plotnikova
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Arya Mohanty
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Bruno B Averbeck
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Kareem Zaghloul
- National Institute of Neurological Disorders and Stroke (NINDS) Intramural Research Program, NIH, Bethesda, MD 20892, USA
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - 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
| | - Kenneth A Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Chris J McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Franciosa F, Acuña MA, Nevian NE, Nevian T. A cellular mechanism contributing to pain-induced analgesia. Pain 2024; 165:2517-2529. [PMID: 38968393 PMCID: PMC11474934 DOI: 10.1097/j.pain.0000000000003315] [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: 09/29/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 07/07/2024]
Abstract
ABSTRACT The anterior cingulate cortex (ACC) plays a crucial role in the perception of pain. It is consistently activated by noxious stimuli and its hyperactivity in chronic pain indicates plasticity in the local neuronal network. However, the way persistent pain effects and modifies different neuronal cell types in the ACC and how this contributes to sensory sensitization is not completely understood. This study confirms the existence of 2 primary subtypes of pyramidal neurons in layer 5 of the rostral, agranular ACC, which we could classify as intratelencephalic (IT) and cortico-subcortical (SC) projecting neurons, similar to other cortical brain areas. Through retrograde labeling, whole-cell patch-clamp recording, and morphological analysis, we thoroughly characterized their different electrophysiological and morphological properties. When examining the effects of peripheral inflammatory pain on these neuronal subtypes, we observed time-dependent plastic changes in excitability. During the acute phase, both subtypes exhibited reduced excitability, which normalized to pre-inflammatory levels after day 7. Daily conditioning with nociceptive stimuli during this period induced an increase in excitability specifically in SC neurons, which was correlated with a decrease in mechanical sensitization. Subsequent inhibition of the activity of SC neurons projecting to the periaqueductal gray with in vivo chemogenetics, resulted in reinstatement of the hypersensitivity. Accordingly, it was sufficient to enhance the excitability of these neurons chemogenetically in the inflammatory pain condition to induce hypoalgesia. These findings suggest a cell type-specific effect on the descending control of nociception and a cellular mechanism for pain-induced analgesia. Furthermore, increased excitability in this neuronal population is hypoalgesic rather than hyperalgesic.
Collapse
Affiliation(s)
| | - Mario A. Acuña
- Department of Physiology, University of Bern, Bern, Switzerland
| | | | - Thomas Nevian
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
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
|
9
|
Luo F, Jiang L, Desai NS, Bai L, Watkins GV, Eldridge MAG, Plotnikova AS, Mohanty A, Cummins AC, Averbeck BB, Talmage DA, Role LW. Comparative Physiology and Morphology of BLA-Projecting NBM/SI Cholinergic Neurons in Mouse and Macaque. J Comp Neurol 2024; 532:e70001. [PMID: 39576005 PMCID: PMC11583843 DOI: 10.1002/cne.70001] [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: 08/13/2024] [Revised: 10/20/2024] [Accepted: 11/06/2024] [Indexed: 11/25/2024]
Abstract
Cholinergic projection neurons of the nucleus basalis and substantia innominata (NBM/SI) densely innervate the basolateral amygdala (BLA) and have been shown to contribute to the encoding of fundamental and life-threatening experiences. Given the vital importance of these circuits in the acquisition and retention of memories that are essential for survival in a changing environment, it is not surprising that the basic anatomical organization of the NBM/SI is well conserved across animal classes as diverse as teleost and mammal. What is not known is the extent to which the physiology and morphology of NBM/SI neurons have also been conserved. To address this issue, we made patch-clamp recordings from NBM/SI neurons in ex vivo slices of two widely divergent mammalian species, mouse and rhesus macaque, focusing our efforts on cholinergic neurons that project to the BLA. We then reconstructed most of these recorded neurons post hoc to characterize neuronal morphology. We found that rhesus macaque BLA-projecting cholinergic neurons were both more intrinsically excitable and less morphologically compact than their mouse homologs. Combining measurements of 18 physiological features and 13 morphological features, we illustrate the extent of the separation. Although macaque and mouse neurons both exhibited considerable within-group diversity and overlapped with each other on multiple individual metrics, a combined morphoelectric analysis demonstrates that they form two distinct neuronal classes. Given the shared purpose of the circuits in which these neurons participate, this finding raises questions about (and offers constraints on) how these distinct classes result in similar behavior.
Collapse
Affiliation(s)
- Feng Luo
- Section on Circuits, Synapses, and Molecular SignalingNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Li Jiang
- Section on Genetics of Neuronal Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Niraj S. Desai
- Section on Circuits, Synapses, and Molecular SignalingNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Li Bai
- Section on Circuits, Synapses, and Molecular SignalingNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Gabrielle V. Watkins
- Section on Circuits, Synapses, and Molecular SignalingNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Mark A. G. Eldridge
- Laboratory of NeuropsychologyNational Institute of Mental Health, National Institutes of HealthBethesdaMarylandUSA
| | - Anya S. Plotnikova
- Laboratory of NeuropsychologyNational Institute of Mental Health, National Institutes of HealthBethesdaMarylandUSA
| | - Arya Mohanty
- Laboratory of NeuropsychologyNational Institute of Mental Health, National Institutes of HealthBethesdaMarylandUSA
| | - Alex C. Cummins
- Laboratory of NeuropsychologyNational Institute of Mental Health, National Institutes of HealthBethesdaMarylandUSA
| | - Bruno B. Averbeck
- Laboratory of NeuropsychologyNational Institute of Mental Health, National Institutes of HealthBethesdaMarylandUSA
| | - David A. Talmage
- Section on Genetics of Neuronal Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Lorna W. Role
- Section on Circuits, Synapses, and Molecular SignalingNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
10
|
Medvediev VV, Cherkasov VG, Marushchenko MO, Vaslovych VV, Tsymbaliuk VI. Giant Fusiform Cells of the Brain: Discovery, Identification, and Probable Functions. CYTOL GENET+ 2024; 58:411-427. [DOI: 10.3103/s0095452724050098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/18/2024] [Accepted: 06/17/2024] [Indexed: 01/05/2025]
|
11
|
Zhao HT, Schmidt ER. Human-specific genetic modifiers of cortical architecture and function. Curr Opin Genet Dev 2024; 88:102241. [PMID: 39111228 PMCID: PMC11547859 DOI: 10.1016/j.gde.2024.102241] [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: 04/04/2024] [Revised: 06/30/2024] [Accepted: 07/23/2024] [Indexed: 09/11/2024]
Abstract
Evolution of the cerebral cortex is thought to have been critical for the emergence of our cognitive abilities. Major features of cortical evolution include increased neuron number and connectivity and altered morpho-electric properties of cortical neurons. Significant progress has been made in identifying human-specific genetic modifiers (HSGMs), some of which are involved in shaping these features of cortical architecture. But how did these evolutionary changes support the emergence of our cognitive abilities? Here, we highlight recent studies aimed at examining the impact of HSGMs on cortical circuit function and behavior. We also discuss the need for greater insight into the link between evolution of cortical architecture and the functional and computational properties of neuronal circuits, as we seek to provide a neurobiological foundation for human cognition.
Collapse
Affiliation(s)
- Hanzhi T Zhao
- Department of Neuroscience, Medical University of South Carolina, Suite 403 BSB, MSC510, 173 Ashley Ave, Charleston, SC 29425, USA
| | - Ewoud Re Schmidt
- Department of Neuroscience, Medical University of South Carolina, Suite 403 BSB, MSC510, 173 Ashley Ave, Charleston, SC 29425, USA.
| |
Collapse
|
12
|
Dembrow NC, Sawchuk S, Dalley R, Opitz-Araya X, Hudson M, Radaelli C, Alfiler L, Walling-Bell S, Bertagnolli D, Goldy J, Johansen N, Miller JA, Nasirova K, Owen SF, Parga-Becerra A, Taskin N, Tieu M, Vumbaco D, Weed N, Wilson J, Lee BR, Smith KA, Sorensen SA, Spain WJ, Lein ES, Perlmutter SI, Ting JT, Kalmbach BE. Areal specializations in the morpho-electric and transcriptomic properties of primate layer 5 extratelencephalic projection neurons. Cell Rep 2024; 43:114718. [PMID: 39277859 PMCID: PMC11488157 DOI: 10.1016/j.celrep.2024.114718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/22/2024] [Accepted: 08/20/2024] [Indexed: 09/17/2024] Open
Abstract
Large-scale analysis of single-cell gene expression has revealed transcriptomically defined cell subclasses present throughout the primate neocortex with gene expression profiles that differ depending upon neocortical region. Here, we test whether the interareal differences in gene expression translate to regional specializations in the physiology and morphology of infragranular glutamatergic neurons by performing Patch-seq experiments in brain slices from the temporal cortex (TCx) and motor cortex (MCx) of the macaque. We confirm that transcriptomically defined extratelencephalically projecting neurons of layer 5 (L5 ET neurons) include retrogradely labeled corticospinal neurons in the MCx and find multiple physiological properties and ion channel genes that distinguish L5 ET from non-ET neurons in both areas. Additionally, while infragranular ET and non-ET neurons retain distinct neuronal properties across multiple regions, there are regional morpho-electric and gene expression specializations in the L5 ET subclass, providing mechanistic insights into the specialized functional architecture of the primate neocortex.
Collapse
Affiliation(s)
- Nikolai C Dembrow
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Epilepsy Center of Excellence, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA.
| | - Scott Sawchuk
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rachel Dalley
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Mark Hudson
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | | | - Lauren Alfiler
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | - Scott F Owen
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Alejandro Parga-Becerra
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - David Vumbaco
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Julia Wilson
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brian R Lee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - William J Spain
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Epilepsy Center of Excellence, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Steve I Perlmutter
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Washington National Primate Research Center, Seattle, WA 98195, USA
| | - Jonathan T Ting
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Allen Institute for Brain Science, Seattle, WA 98109, USA; Washington National Primate Research Center, Seattle, WA 98195, USA
| | - Brian E Kalmbach
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Allen Institute for Brain Science, Seattle, WA 98109, USA.
| |
Collapse
|
13
|
Luo F, Jiang L, Desai NS, Bai L, Watkins GV, Eldridge MAG, Plotnikova A, Mohanty A, Cummins AC, Averbeck BB, Talmage DA, Role LW. Comparative physiology and morphology of BLA-projecting NBM/SI cholinergic neurons in mouse and macaque. RESEARCH SQUARE 2024:rs.3.rs-4824445. [PMID: 39149491 PMCID: PMC11326416 DOI: 10.21203/rs.3.rs-4824445/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Cholinergic projection neurons of the nucleus basalis and substantia innominata (NBM/SI) densely innervate the basolateral amygdala (BLA) and have been shown to contribute to the encoding of fundamental and life-threatening experiences. Given the vital importance of these circuits in the acquisition and retention of memories that are essential for survival in a changing environment, it is not surprising that the basic anatomical organization of the NBM/SI is well conserved across animal classes as diverse as teleost and mammal. What is not known is the extent to which the physiology and morphology of NBM/SI neurons have also been conserved. To address this issue, we made patch-clamp recordings from NBM/SI neurons in ex vivo slices of two widely divergent mammalian species, mouse and rhesus macaque, focusing our efforts on cholinergic neurons that project to the BLA. We then reconstructed most of these recorded neurons post hoc to characterize neuronal morphology. We found that rhesus macaque BLA-projecting cholinergic neurons were both more intrinsically excitable and less morphologically compact than their mouse homologs. Combining measurements of 18 physiological features and 13 morphological features, we illustrate the extent of the separation. Although macaque and mouse neurons both exhibited considerable within-group diversity and overlapped with each other on multiple individual metrics, a combined morpho-electric analysis demonstrates that they form two distinct neuronal classes. Given the shared purpose of the circuits in which these neurons participate, this finding raises questions about (and offers constraints on) how these distinct classes result in similar behavior.
Collapse
Affiliation(s)
- Feng Luo
- Section on Circuits, Synapses, and Molecular Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Li Jiang
- Section on Genetics of Neuronal Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Niraj S. Desai
- Section on Circuits, Synapses, and Molecular Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Li Bai
- Section on Circuits, Synapses, and Molecular Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Gabrielle V. Watkins
- Section on Circuits, Synapses, and Molecular Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Mark A. G. Eldridge
- Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, 20892, MD, USA
| | - Anya Plotnikova
- Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, 20892, MD, USA
| | - Arya Mohanty
- Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, 20892, MD, USA
| | - Alex C. Cummins
- Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, 20892, MD, USA
| | - Bruno B. Averbeck
- Laboratory of Neuropsychology, National Institute of Mental Health, Bethesda, 20892, MD, USA
| | - David A. Talmage
- Section on Genetics of Neuronal Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Lorna W. Role
- Section on Circuits, Synapses, and Molecular Signaling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, 20892, MD, USA
| |
Collapse
|
14
|
Mahon S. Variation and convergence in the morpho-functional properties of the mammalian neocortex. Front Syst Neurosci 2024; 18:1413780. [PMID: 38966330 PMCID: PMC11222651 DOI: 10.3389/fnsys.2024.1413780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
Man's natural inclination to classify and hierarchize the living world has prompted neurophysiologists to explore possible differences in brain organisation between mammals, with the aim of understanding the diversity of their behavioural repertoires. But what really distinguishes the human brain from that of a platypus, an opossum or a rodent? In this review, we compare the structural and electrical properties of neocortical neurons in the main mammalian radiations and examine their impact on the functioning of the networks they form. We discuss variations in overall brain size, number of neurons, length of their dendritic trees and density of spines, acknowledging their increase in humans as in most large-brained species. Our comparative analysis also highlights a remarkable consistency, particularly pronounced in marsupial and placental mammals, in the cell typology, intrinsic and synaptic electrical properties of pyramidal neuron subtypes, and in their organisation into functional circuits. These shared cellular and network characteristics contribute to the emergence of strikingly similar large-scale physiological and pathological brain dynamics across a wide range of species. These findings support the existence of a core set of neural principles and processes conserved throughout mammalian evolution, from which a number of species-specific adaptations appear, likely allowing distinct functional needs to be met in a variety of environmental contexts.
Collapse
Affiliation(s)
- Séverine Mahon
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, France
| |
Collapse
|
15
|
Yagishita H, Sasaki T. Integrating physiological and transcriptomic analyses at the single-neuron level. Neurosci Res 2024:S0168-0102(24)00065-8. [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] [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; Department of Neuropharmacology, Tohoku University School of Medicine, 4-1 Seiryo-machi, Aoba-Ku, Sendai 980-8575, Japan.
| |
Collapse
|
16
|
Kampmann M. Molecular and cellular mechanisms of selective vulnerability in neurodegenerative diseases. Nat Rev Neurosci 2024; 25:351-371. [PMID: 38575768 DOI: 10.1038/s41583-024-00806-0] [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: 03/01/2024] [Indexed: 04/06/2024]
Abstract
The selective vulnerability of specific neuronal subtypes is a hallmark of neurodegenerative diseases. In this Review, I summarize our current understanding of the brain regions and cell types that are selectively vulnerable in different neurodegenerative diseases and describe the proposed underlying cell-autonomous and non-cell-autonomous mechanisms. I highlight how recent methodological innovations - including single-cell transcriptomics, CRISPR-based screens and human cell-based models of disease - are enabling new breakthroughs in our understanding of selective vulnerability. An understanding of the molecular mechanisms that determine selective vulnerability and resilience would shed light on the key processes that drive neurodegeneration and point to potential therapeutic strategies to protect vulnerable cell populations.
Collapse
Affiliation(s)
- Martin Kampmann
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
17
|
Zhou Y, He W, Hou W, Zhu Y. Pianno: a probabilistic framework automating semantic annotation for spatial transcriptomics. Nat Commun 2024; 15:2848. [PMID: 38565531 PMCID: PMC11271244 DOI: 10.1038/s41467-024-47152-4] [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: 09/12/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Spatial transcriptomics has revolutionized the study of gene expression within tissues, while preserving spatial context. However, annotating spatial spots' biological identity remains a challenge. To tackle this, we introduce Pianno, a Bayesian framework automating structural semantics annotation based on marker genes. Comprehensive evaluations underscore Pianno's remarkable prowess in precisely annotating a wide array of spatial semantics, ranging from diverse anatomical structures to intricate tumor microenvironments, as well as in estimating cell type distributions, across data generated from various spatial transcriptomics platforms. Furthermore, Pianno, in conjunction with clustering approaches, uncovers a region- and species-specific excitatory neuron subtype in the deep layer 3 of the human neocortex, shedding light on cellular evolution in the human neocortex. Overall, Pianno equips researchers with a robust and efficient tool for annotating diverse biological structures, offering new perspectives on spatial transcriptomics data.
Collapse
Affiliation(s)
- Yuqiu Zhou
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei He
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Weizhen Hou
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Shen Y, Shao M, Hao ZZ, Huang M, Xu N, Liu S. Multimodal Nature of the Single-cell Primate Brain Atlas: Morphology, Transcriptome, Electrophysiology, and Connectivity. Neurosci Bull 2024; 40:517-532. [PMID: 38194157 PMCID: PMC11003949 DOI: 10.1007/s12264-023-01160-4] [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: 03/22/2023] [Accepted: 09/23/2023] [Indexed: 01/10/2024] Open
Abstract
Primates exhibit complex brain structures that augment cognitive function. The neocortex fulfills high-cognitive functions through billions of connected neurons. These neurons have distinct transcriptomic, morphological, and electrophysiological properties, and their connectivity principles vary. These features endow the primate brain atlas with a multimodal nature. The recent integration of next-generation sequencing with modified patch-clamp techniques is revolutionizing the way to census the primate neocortex, enabling a multimodal neuronal atlas to be established in great detail: (1) single-cell/single-nucleus RNA-seq technology establishes high-throughput transcriptomic references, covering all major transcriptomic cell types; (2) patch-seq links the morphological and electrophysiological features to the transcriptomic reference; (3) multicell patch-clamp delineates the principles of local connectivity. Here, we review the applications of these technologies in the primate neocortex and discuss the current advances and tentative gaps for a comprehensive understanding of the primate neocortex.
Collapse
Affiliation(s)
- Yuhui Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Mingting Shao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Zhao-Zhe Hao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Mengyao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Nana Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, 510080, China.
| |
Collapse
|
19
|
Khodosevich K, Dragicevic K, Howes O. Drug targeting in psychiatric disorders - how to overcome the loss in translation? Nat Rev Drug Discov 2024; 23:218-231. [PMID: 38114612 DOI: 10.1038/s41573-023-00847-7] [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: 11/03/2023] [Indexed: 12/21/2023]
Abstract
In spite of major efforts and investment in development of psychiatric drugs, many clinical trials have failed in recent decades, and clinicians still prescribe drugs that were discovered many years ago. Although multiple reasons have been discussed for the drug development deadlock, we focus here on one of the major possible biological reasons: differences between the characteristics of drug targets in preclinical models and the corresponding targets in patients. Importantly, based on technological advances in single-cell analysis, we propose here a framework for the use of available and newly emerging knowledge from single-cell and spatial omics studies to evaluate and potentially improve the translational predictivity of preclinical models before commencing preclinical and, in particular, clinical studies. We believe that these recommendations will improve preclinical models and the ability to assess drugs in clinical trials, reducing failure rates in expensive late-stage trials and ultimately benefitting psychiatric drug discovery and development.
Collapse
Affiliation(s)
- Konstantin Khodosevich
- Biotech Research and Innovation Centre, Faculty of Health, University of Copenhagen, Copenhagen, Denmark.
| | - Katarina Dragicevic
- Biotech Research and Innovation Centre, Faculty of Health, University of Copenhagen, Copenhagen, Denmark
| | - Oliver Howes
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
20
|
Guet-McCreight A, Chameh HM, Mazza F, Prevot TD, Valiante TA, Sibille E, Hay E. In-silico testing of new pharmacology for restoring inhibition and human cortical function in depression. Commun Biol 2024; 7:225. [PMID: 38396202 PMCID: PMC10891083 DOI: 10.1038/s42003-024-05907-1] [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: 08/30/2023] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Reduced inhibition by somatostatin-expressing interneurons is associated with depression. Administration of positive allosteric modulators of α5 subunit-containing GABAA receptor (α5-PAM) that selectively target this lost inhibition exhibit antidepressant and pro-cognitive effects in rodent models of chronic stress. However, the functional effects of α5-PAM on the human brain in vivo are unknown, and currently cannot be assessed experimentally. We modeled the effects of α5-PAM on tonic inhibition as measured in human neurons, and tested in silico α5-PAM effects on detailed models of human cortical microcircuits in health and depression. We found that α5-PAM effectively recovered impaired cortical processing as quantified by stimulus detection metrics, and also recovered the power spectral density profile of the microcircuit EEG signals. We performed an α5-PAM dose-response and identified simulated EEG biomarker candidates. Our results serve to de-risk and facilitate α5-PAM translation and provide biomarkers in non-invasive brain signals for monitoring target engagement and drug efficacy.
Collapse
Affiliation(s)
- Alexandre Guet-McCreight
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | | | - Frank Mazza
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Thomas D Prevot
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Taufik A Valiante
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
- Center for Advancing Neurotechnological Innovation to Application, Toronto, ON, Canada
- Max Planck-University of Toronto Center for Neural Science and Technology, Toronto, ON, Canada
| | - Etienne Sibille
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Etay Hay
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
21
|
Camunas-Soler J. Integrating single-cell transcriptomics with cellular phenotypes: cell morphology, Ca 2+ imaging and electrophysiology. Biophys Rev 2024; 16:89-107. [PMID: 38495444 PMCID: PMC10937895 DOI: 10.1007/s12551-023-01174-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/29/2023] [Indexed: 03/19/2024] Open
Abstract
I review recent technological advancements in coupling single-cell transcriptomics with cellular phenotypes including morphology, calcium signaling, and electrophysiology. Single-cell RNA sequencing (scRNAseq) has revolutionized cell type classifications by capturing the transcriptional diversity of cells. A new wave of methods to integrate scRNAseq and biophysical measurements is facilitating the linkage of transcriptomic data to cellular function, which provides physiological insight into cellular states. I briefly discuss critical factors of these phenotypical characterizations such as timescales, information content, and analytical tools. Dedicated sections focus on the integration with cell morphology, calcium imaging, and electrophysiology (patch-seq), emphasizing their complementary roles. I discuss their application in elucidating cellular states, refining cell type classifications, and uncovering functional differences in cell subtypes. To illustrate the practical applications and benefits of these methods, I highlight their use in tissues with excitable cell-types such as the brain, pancreatic islets, and the retina. The potential of combining functional phenotyping with spatial transcriptomics for a detailed mapping of cell phenotypes in situ is explored. Finally, I discuss open questions and future perspectives, emphasizing the need for a shift towards broader accessibility through increased throughput.
Collapse
Affiliation(s)
- Joan Camunas-Soler
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| |
Collapse
|
22
|
Bahl E, Chatterjee S, Mukherjee U, Elsadany M, Vanrobaeys Y, Lin LC, McDonough M, Resch J, Giese KP, Abel T, Michaelson JJ. Using deep learning to quantify neuronal activation from single-cell and spatial transcriptomic data. Nat Commun 2024; 15:779. [PMID: 38278804 PMCID: PMC10817898 DOI: 10.1038/s41467-023-44503-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/15/2023] [Indexed: 01/28/2024] Open
Abstract
Neuronal activity-dependent transcription directs molecular processes that regulate synaptic plasticity, brain circuit development, behavioral adaptation, and long-term memory. Single cell RNA-sequencing technologies (scRNAseq) are rapidly developing and allow for the interrogation of activity-dependent transcription at cellular resolution. Here, we present NEUROeSTIMator, a deep learning model that integrates transcriptomic signals to estimate neuronal activation in a way that we demonstrate is associated with Patch-seq electrophysiological features and that is robust against differences in species, cell type, and brain region. We demonstrate this method's ability to accurately detect neuronal activity in previously published studies of single cell activity-induced gene expression. Further, we applied our model in a spatial transcriptomic study to identify unique patterns of learning-induced activity across different brain regions in male mice. Altogether, our findings establish NEUROeSTIMator as a powerful and broadly applicable tool for measuring neuronal activation, whether as a critical covariate or a primary readout of interest.
Collapse
Affiliation(s)
- Ethan Bahl
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, USA
| | - Snehajyoti Chatterjee
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Utsav Mukherjee
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA
| | - Muhammad Elsadany
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, USA
| | - Yann Vanrobaeys
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Li-Chun Lin
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Miriam McDonough
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Molecular Medicine, University of Iowa, Iowa City, IA, USA
| | - Jon Resch
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
| | - K Peter Giese
- Department of Basic and Clinical Neuroscience, King's College London, London, UK
| | - Ted Abel
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Jacob J Michaelson
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA.
- Department of Communication Sciences & Disorders, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
23
|
Wallace JL, Pollen AA. Human neuronal maturation comes of age: cellular mechanisms and species differences. Nat Rev Neurosci 2024; 25:7-29. [PMID: 37996703 DOI: 10.1038/s41583-023-00760-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 11/25/2023]
Abstract
The delayed and prolonged postmitotic maturation of human neurons, compared with neurons from other species, may contribute to human-specific cognitive abilities and neurological disorders. Here we review the mechanisms of neuronal maturation, applying lessons from model systems to understand the specific features of protracted human cortical maturation and species differences. We cover cell-intrinsic features of neuronal maturation, including transcriptional, epigenetic and metabolic mechanisms, as well as cell-extrinsic features, including the roles of activity and synapses, the actions of glial cells and the contribution of the extracellular matrix. We discuss evidence for species differences in biochemical reaction rates, the proposed existence of an epigenetic maturation clock and the contributions of both general and modular mechanisms to species-specific maturation timing. Finally, we suggest approaches to measure, improve and accelerate the maturation of human neurons in culture, examine crosstalk and interactions among these different aspects of maturation and propose conceptual models to guide future studies.
Collapse
Affiliation(s)
- Jenelle L Wallace
- 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.
| | - 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.
| |
Collapse
|
24
|
Libé-Philippot B, Lejeune A, Wierda K, Louros N, Erkol E, Vlaeminck I, Beckers S, Gaspariunaite V, Bilheu A, Konstantoulea K, Nyitrai H, De Vleeschouwer M, Vennekens KM, Vidal N, Bird TW, Soto DC, Jaspers T, Dewilde M, Dennis MY, Rousseau F, Comoletti D, Schymkowitz J, Theys T, de Wit J, Vanderhaeghen P. LRRC37B is a human modifier of voltage-gated sodium channels and axon excitability in cortical neurons. Cell 2023; 186:5766-5783.e25. [PMID: 38134874 PMCID: PMC10754148 DOI: 10.1016/j.cell.2023.11.028] [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: 12/02/2022] [Revised: 06/28/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023]
Abstract
The enhanced cognitive abilities characterizing the human species result from specialized features of neurons and circuits. Here, we report that the hominid-specific gene LRRC37B encodes a receptor expressed in human cortical pyramidal neurons (CPNs) and selectively localized to the axon initial segment (AIS), the subcellular compartment triggering action potentials. Ectopic expression of LRRC37B in mouse CPNs in vivo leads to reduced intrinsic excitability, a distinctive feature of some classes of human CPNs. Molecularly, LRRC37B binds to the secreted ligand FGF13A and to the voltage-gated sodium channel (Nav) β-subunit SCN1B. LRRC37B concentrates inhibitory effects of FGF13A on Nav channel function, thereby reducing excitability, specifically at the AIS level. Electrophysiological recordings in adult human cortical slices reveal lower neuronal excitability in human CPNs expressing LRRC37B. LRRC37B thus acts as a species-specific modifier of human neuron excitability, linking human genome and cell evolution, with important implications for human brain function and diseases.
Collapse
Affiliation(s)
- Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Amélie Lejeune
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Nikolaos Louros
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Emir Erkol
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Ine Vlaeminck
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Sofie Beckers
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Vaiva Gaspariunaite
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Angéline Bilheu
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium
| | - Katerina Konstantoulea
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Hajnalka Nyitrai
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Matthias De Vleeschouwer
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Kristel M Vennekens
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Niels Vidal
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium
| | - Thomas W Bird
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Daniela C Soto
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Tom Jaspers
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Maarten Dewilde
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Megan Y Dennis
- Genome Center, MIND Institute, and Department of Biochemistry & Molecular Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Frederic Rousseau
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Davide Comoletti
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Child Health Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Joost Schymkowitz
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Cellular and Molecular Medicine, KUL, 3000 Leuven, Belgium
| | - Tom Theys
- KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; Research Group Experimental Neurosurgery and Neuroanatomy, KUL, 3000 Leuven, Belgium
| | - Joris de Wit
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium.
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KUL, Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), 1070 Brussels, Belgium.
| |
Collapse
|
25
|
Shao M, Zhang W, Li Y, Tang L, Hao ZZ, Liu S. Patch-seq: Advances and Biological Applications. Cell Mol Neurobiol 2023; 44:8. [PMID: 38123823 DOI: 10.1007/s10571-023-01436-3] [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: 05/11/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023]
Abstract
Multimodal analysis of gene-expression patterns, electrophysiological properties, and morphological phenotypes at the single-cell/single-nucleus level has been arduous because of the diversity and complexity of neurons. The emergence of Patch-sequencing (Patch-seq) directly links transcriptomics, morphology, and electrophysiology, taking neuroscience research to a multimodal era. In this review, we summarized the development of Patch-seq and recent applications in the cortex, hippocampus, and other nervous systems. Through generating multimodal cell type atlases, targeting specific cell populations, and correlating transcriptomic data with phenotypic information, Patch-seq has provided new insight into outstanding questions in neuroscience. We highlight the challenges and opportunities of Patch-seq in neuroscience and hope to shed new light on future neuroscience research.
Collapse
Affiliation(s)
- Mingting Shao
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Wei Zhang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Ye Li
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Lei Tang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Zhao-Zhe Hao
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, 510080, China.
| |
Collapse
|
26
|
Pelkey KA, Vargish GA, Pellegrini LV, Calvigioni D, Chapeton J, Yuan X, Hunt S, Cummins AC, Eldridge MAG, Pickel J, Chittajallu R, Averbeck BB, Tóth K, Zaghloul K, McBain CJ. Evolutionary conservation of hippocampal mossy fiber synapse properties. Neuron 2023; 111:3802-3818.e5. [PMID: 37776852 PMCID: PMC10841147 DOI: 10.1016/j.neuron.2023.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/03/2023] [Accepted: 09/06/2023] [Indexed: 10/02/2023]
Abstract
Various specialized structural/functional properties are considered essential for contextual memory encoding by hippocampal mossy fiber (MF) synapses. Although investigated to exquisite detail in model organisms, synapses, including MFs, have undergone minimal functional interrogation in humans. To determine the translational relevance of rodent findings, we evaluated MF properties within human tissue resected to treat epilepsy. Human MFs exhibit remarkably similar hallmark features to rodents, including AMPA receptor-dominated synapses with small contributions from NMDA and kainate receptors, large dynamic range with strong frequency facilitation, NMDA receptor-independent presynaptic long-term potentiation, and strong cyclic AMP (cAMP) sensitivity of release. Array tomography confirmed the evolutionary conservation of MF ultrastructure. The astonishing congruence of rodent and human MF core features argues that the basic MF properties delineated in animal models remain critical to human MF function. Finally, a selective deficit in GABAergic inhibitory tone onto human MF postsynaptic targets suggests that unrestrained detonator excitatory drive contributes to epileptic circuit hyperexcitability.
Collapse
Affiliation(s)
- Kenneth A Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Geoffrey A Vargish
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Leonardo V Pellegrini
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Brain and Mind Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Daniela Calvigioni
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Julio Chapeton
- National Institute of Neurological Disorders and Stroke Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiaoqing Yuan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven Hunt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alex C Cummins
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark A G Eldridge
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - James Pickel
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bruno B Averbeck
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katalin Tóth
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Brain and Mind Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Kareem Zaghloul
- National Institute of Neurological Disorders and Stroke Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chris J McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Herrera B, Sajad A, Errington SP, Schall JD, Riera JJ. Cortical origin of theta error signals. Cereb Cortex 2023; 33:11300-11319. [PMID: 37804250 PMCID: PMC10690871 DOI: 10.1093/cercor/bhad367] [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: 06/23/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/09/2023] Open
Abstract
A multi-scale approach elucidated the origin of the error-related-negativity (ERN), with its associated theta-rhythm, and the post-error-positivity (Pe) in macaque supplementary eye field (SEF). Using biophysical modeling, synaptic inputs to a subpopulation of layer-3 (L3) and layer-5 (L5) pyramidal cells (PCs) were optimized to reproduce error-related spiking modulation and inter-spike intervals. The intrinsic dynamics of dendrites in L5 but not L3 error PCs generate theta rhythmicity with random phases. Saccades synchronized the phases of the theta-rhythm, which was magnified on errors. Contributions from error PCs to the laminar current source density (CSD) observed in SEF were negligible and could not explain the observed association between error-related spiking modulation in L3 PCs and scalp-EEG. CSD from recorded laminar field potentials in SEF was comprised of multipolar components, with monopoles indicating strong electro-diffusion, dendritic/axonal electrotonic current leakage outside SEF, or violations of the model assumptions. Our results also demonstrate the involvement of secondary cortical regions, in addition to SEF, particularly for the later Pe component. The dipolar component from the observed CSD paralleled the ERN dynamics, while the quadrupolar component paralleled the Pe. These results provide the most advanced explanation to date of the cellular mechanisms generating the ERN.
Collapse
Affiliation(s)
- Beatriz Herrera
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, United States
| | - Amirsaman Sajad
- Department of Psychology, Vanderbilt Vision Research Center, Center for Integrative & Cognitive Neuroscience, Vanderbilt University, Nashville, TN 37203, United States
| | - Steven P Errington
- Department of Psychology, Vanderbilt Vision Research Center, Center for Integrative & Cognitive Neuroscience, Vanderbilt University, Nashville, TN 37203, United States
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Jeffrey D Schall
- Centre for Vision Research, Vision: Science to Applications Program, Departments of Biology and Psychology, York University, Toronto, ON M3J 1P3, Canada
| | - Jorge J Riera
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, United States
| |
Collapse
|
28
|
Chartrand T, Dalley R, Close J, Goriounova NA, Lee BR, Mann R, Miller JA, Molnar G, Mukora A, Alfiler L, Baker K, Bakken TE, Berg J, Bertagnolli D, Braun T, Brouner K, Casper T, Csajbok EA, Dee N, Egdorf T, Enstrom R, Galakhova AA, Gary A, Gelfand E, Goldy J, Hadley K, Heistek TS, Hill D, Jorstad N, Kim L, Kocsis AK, Kruse L, Kunst M, Leon G, Long B, Mallory M, McGraw M, McMillen D, Melief EJ, Mihut N, Ng L, Nyhus J, Oláh G, Ozsvár A, Omstead V, Peterfi Z, Pom A, Potekhina L, Rajanbabu R, Rozsa M, Ruiz A, Sandle J, Sunkin SM, Szots I, Tieu M, Toth M, Trinh J, Vargas S, Vumbaco D, Williams G, Wilson J, Yao Z, Barzo P, Cobbs C, Ellenbogen RG, Esposito L, Ferreira M, Gouwens NW, Grannan B, Gwinn RP, Hauptman JS, Jarsky T, Keene CD, Ko AL, Koch C, Ojemann JG, Patel A, Ruzevick J, Silbergeld DL, Smith K, Sorensen SA, Tasic B, Ting JT, Waters J, de Kock CPJ, Mansvelder HD, Tamas G, Zeng H, Kalmbach B, Lein ES. Morphoelectric and transcriptomic divergence of the layer 1 interneuron repertoire in human versus mouse neocortex. Science 2023; 382:eadf0805. [PMID: 37824667 DOI: 10.1126/science.adf0805] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 09/09/2023] [Indexed: 10/14/2023]
Abstract
Neocortical layer 1 (L1) is a site of convergence between pyramidal-neuron dendrites and feedback axons where local inhibitory signaling can profoundly shape cortical processing. Evolutionary expansion of human neocortex is marked by distinctive pyramidal neurons with extensive L1 branching, but whether L1 interneurons are similarly diverse is underexplored. Using Patch-seq recordings from human neurosurgical tissue, we identified four transcriptomic subclasses with mouse L1 homologs, along with distinct subtypes and types unmatched in mouse L1. Subclass and subtype comparisons showed stronger transcriptomic differences in human L1 and were correlated with strong morphoelectric variability along dimensions distinct from mouse L1 variability. Accompanied by greater layer thickness and other cytoarchitecture changes, these findings suggest that L1 has diverged in evolution, reflecting the demands of regulating the expanded human neocortical circuit.
Collapse
Affiliation(s)
| | | | - Jennie Close
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Natalia A Goriounova
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, Netherlands
| | - Brian R Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Rusty Mann
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Gabor Molnar
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | - Alice Mukora
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Jim Berg
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Eva Adrienn Csajbok
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tom Egdorf
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Anna A Galakhova
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, Netherlands
| | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Tim S Heistek
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, Netherlands
| | - DiJon Hill
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nik Jorstad
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lisa Kim
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Agnes Katalin Kocsis
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Brian Long
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Medea McGraw
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Erica J Melief
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Norbert Mihut
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | - Lindsay Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Julie Nyhus
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Gáspár Oláh
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | - Attila Ozsvár
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | | | - Zoltan Peterfi
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | - Alice Pom
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Marton Rozsa
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | | | - Joanna Sandle
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | | | - Ildiko Szots
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Martin Toth
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | | | - Sara Vargas
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Julia Wilson
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Pal Barzo
- Department of Neurosurgery, University of Szeged, Szeged, Hungary
| | | | | | | | - Manuel Ferreira
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | | | - Benjamin Grannan
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | | | - Jason S Hauptman
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Tim Jarsky
- Allen Institute for Brain Science, Seattle, WA, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Andrew L Ko
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | | | - Jeffrey G Ojemann
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Anoop Patel
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Jacob Ruzevick
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Daniel L Silbergeld
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | | | | | | | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Christiaan P J de Kock
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, Netherlands
| | - Huib D Mansvelder
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, Netherlands
| | - Gabor Tamas
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged, Szeged, Hungary
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Brian Kalmbach
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| |
Collapse
|
29
|
Lee BR, Dalley R, Miller JA, Chartrand T, Close J, Mann R, Mukora A, Ng L, Alfiler L, Baker K, Bertagnolli D, Brouner K, Casper T, Csajbok E, Donadio N, Driessens SLW, Egdorf T, Enstrom R, Galakhova AA, Gary A, Gelfand E, Goldy J, Hadley K, Heistek TS, Hill D, Hou WH, Johansen N, Jorstad N, Kim L, Kocsis AK, Kruse L, Kunst M, León G, Long B, Mallory M, Maxwell M, McGraw M, McMillen D, Melief EJ, Molnar G, Mortrud MT, Newman D, Nyhus J, Opitz-Araya X, Ozsvár A, Pham T, Pom A, Potekhina L, Rajanbabu R, Ruiz A, Sunkin SM, Szöts I, Taskin N, Thyagarajan B, Tieu M, Trinh J, Vargas S, Vumbaco D, Waleboer F, Walling-Bell S, Weed N, Williams G, Wilson J, Yao S, Zhou T, Barzó P, Bakken T, Cobbs C, Dee N, Ellenbogen RG, Esposito L, Ferreira M, Gouwens NW, Grannan B, Gwinn RP, Hauptman JS, Hodge R, Jarsky T, Keene CD, Ko AL, Korshoej AR, Levi BP, Meier K, Ojemann JG, Patel A, Ruzevick J, Silbergeld DL, Smith K, Sørensen JC, Waters J, Zeng H, Berg J, Capogna M, Goriounova NA, Kalmbach B, de Kock CPJ, Mansvelder HD, Sorensen SA, Tamas G, Lein ES, Ting JT. Signature morphoelectric properties of diverse GABAergic interneurons in the human neocortex. Science 2023; 382:eadf6484. [PMID: 37824669 DOI: 10.1126/science.adf6484] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 09/08/2023] [Indexed: 10/14/2023]
Abstract
Human cortex transcriptomic studies have revealed a hierarchical organization of γ-aminobutyric acid-producing (GABAergic) neurons from subclasses to a high diversity of more granular types. Rapid GABAergic neuron viral genetic labeling plus Patch-seq (patch-clamp electrophysiology plus single-cell RNA sequencing) sampling in human brain slices was used to reliably target and analyze GABAergic neuron subclasses and individual transcriptomic types. This characterization elucidated transitions between PVALB and SST subclasses, revealed morphological heterogeneity within an abundant transcriptomic type, identified multiple spatially distinct types of the primate-specialized double bouquet cells (DBCs), and shed light on cellular differences between homologous mouse and human neocortical GABAergic neuron types. These results highlight the importance of multimodal phenotypic characterization for refinement of emerging transcriptomic cell type taxonomies and for understanding conserved and specialized cellular properties of human brain cell types.
Collapse
Affiliation(s)
- Brian R Lee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rachel Dalley
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Thomas Chartrand
- Allen Institute for Brain Science, Seattle, WA 98109, USA
- Allen Institute for Neural Dynamics, Seattle, WA 98109, USA
| | - Jennie Close
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rusty Mann
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Alice Mukora
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Lindsay Ng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Lauren Alfiler
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Krissy Brouner
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tamara Casper
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Eva Csajbok
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, 6726 Szeged, Hungary
| | | | - Stan L W Driessens
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, 1081 HV, Netherlands
| | - Tom Egdorf
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rachel Enstrom
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Anna A Galakhova
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, 1081 HV, Netherlands
| | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Emily Gelfand
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kristen Hadley
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tim S Heistek
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, 1081 HV, Netherlands
| | - Dijon Hill
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Wen-Hsien Hou
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | | | - Nik Jorstad
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Lisa Kim
- Allen Institute for Brain Science, Seattle, WA 98109, USA
- Allen Institute for Neural Dynamics, Seattle, WA 98109, USA
| | - Agnes Katalin Kocsis
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, 6726 Szeged, Hungary
| | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Kunst
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Gabriela León
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brian Long
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Medea McGraw
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Erica J Melief
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Gabor Molnar
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, 6726 Szeged, Hungary
| | | | - Dakota Newman
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Julie Nyhus
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Attila Ozsvár
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | | | - Alice Pom
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Ram Rajanbabu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Augustin Ruiz
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Susan M Sunkin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ildikó Szöts
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, 6726 Szeged, Hungary
| | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jessica Trinh
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Sara Vargas
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - David Vumbaco
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Femke Waleboer
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, 1081 HV, Netherlands
| | | | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Grace Williams
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Julia Wilson
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Shenqin Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Thomas Zhou
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Pál Barzó
- Department of Neurosurgery, University of Szeged, 6725 Szeged, Hungary
| | - Trygve Bakken
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Charles Cobbs
- Swedish Neuroscience Institute, Seattle, WA 98122, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Richard G Ellenbogen
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Luke Esposito
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Manuel Ferreira
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | | | - Benjamin Grannan
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Ryder P Gwinn
- Swedish Neuroscience Institute, Seattle, WA 98122, USA
| | - Jason S Hauptman
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Rebecca Hodge
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tim Jarsky
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Andrew L Ko
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | | | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kaare Meier
- Department of Neurosurgery, Aarhus University Hospital, 8200 Aarhus, Denmark
- Department of Anesthesiology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Jeffrey G Ojemann
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Anoop Patel
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Jacob Ruzevick
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Daniel L Silbergeld
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Kimberly Smith
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jens Christian Sørensen
- Department of Neurosurgery, Aarhus University Hospital, 8200 Aarhus, Denmark
- Center for Experimental Neuroscience, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA 98109, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jim Berg
- Allen Institute for Brain Science, Seattle, WA 98109, USA
- Allen Institute for Neural Dynamics, Seattle, WA 98109, USA
| | - Marco Capogna
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Natalia A Goriounova
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, 1081 HV, Netherlands
| | - Brian Kalmbach
- Allen Institute for Brain Science, Seattle, WA 98109, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Christiaan P J de Kock
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, 1081 HV, Netherlands
| | - Huib D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit, Amsterdam, 1081 HV, Netherlands
| | | | - Gabor Tamas
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy, and Neuroscience, University of Szeged, 6726 Szeged, Hungary
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA 98109, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
30
|
Han X, Guo S, Ji N, Li T, Liu J, Ye X, Wang Y, Yun Z, Xiong F, Rong J, Liu D, Ma H, Wang Y, Huang Y, Zhang P, Wu W, Ding L, Hawrylycz M, Lein E, Ascoli GA, Xie W, Liu L, Zhang L, Peng H. Whole human-brain mapping of single cortical neurons for profiling morphological diversity and stereotypy. SCIENCE ADVANCES 2023; 9:eadf3771. [PMID: 37824619 PMCID: PMC10569712 DOI: 10.1126/sciadv.adf3771] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 04/18/2023] [Indexed: 10/14/2023]
Abstract
Quantifying neuron morphology and distribution at the whole-brain scale is essential to understand the structure and diversity of cell types. It is exceedingly challenging to reuse recent technologies of single-cell labeling and whole-brain imaging to study human brains. We propose adaptive cell tomography (ACTomography), a low-cost, high-throughput, and high-efficacy tomography approach, based on adaptive targeting of individual cells. We established a platform to inject dyes into cortical neurons in surgical tissues of 18 patients with brain tumors or other conditions and one donated fresh postmortem brain. We collected three-dimensional images of 1746 cortical neurons, of which 852 neurons were reconstructed to quantify local dendritic morphology, and mapped to standard atlases. In our data, human neurons are more diverse across brain regions than by subject age or gender. The strong stereotypy within cohorts of brain regions allows generating a statistical tensor field of neuron morphology to characterize anatomical modularity of a human brain.
Collapse
Affiliation(s)
- Xiaofeng Han
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Shuxia Guo
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Tian Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jian Liu
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Xiangqiao Ye
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Yi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhixi Yun
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Feng Xiong
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Jing Rong
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Di Liu
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Hui Ma
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Yujin Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yue Huang
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenhao Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liya Ding
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | | | - Ed Lein
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Giorgio A. Ascoli
- Center for Neural Informatics, Krasnow Institute for Advanced Studies and Bioengineering Department, College of Engineering and Computing, George Mason University, Fairfax, VA, USA
| | - Wei Xie
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Lijuan Liu
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Hanchuan Peng
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| |
Collapse
|
31
|
Chameh HM, Falby M, Movahed M, Arbabi K, Rich S, Zhang L, Lefebvre J, Tripathy SJ, De Pittà M, Valiante TA. Distinctive biophysical features of human cell-types: insights from studies of neurosurgically resected brain tissue. Front Synaptic Neurosci 2023; 15:1250834. [PMID: 37860223 PMCID: PMC10584155 DOI: 10.3389/fnsyn.2023.1250834] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/21/2023] [Indexed: 10/21/2023] Open
Abstract
Electrophysiological characterization of live human tissue from epilepsy patients has been performed for many decades. Although initially these studies sought to understand the biophysical and synaptic changes associated with human epilepsy, recently, it has become the mainstay for exploring the distinctive biophysical and synaptic features of human cell-types. Both epochs of these human cellular electrophysiological explorations have faced criticism. Early studies revealed that cortical pyramidal neurons obtained from individuals with epilepsy appeared to function "normally" in comparison to neurons from non-epilepsy controls or neurons from other species and thus there was little to gain from the study of human neurons from epilepsy patients. On the other hand, contemporary studies are often questioned for the "normalcy" of the recorded neurons since they are derived from epilepsy patients. In this review, we discuss our current understanding of the distinct biophysical features of human cortical neurons and glia obtained from tissue removed from patients with epilepsy and tumors. We then explore the concept of within cell-type diversity and its loss (i.e., "neural homogenization"). We introduce neural homogenization to help reconcile the epileptogenicity of seemingly "normal" human cortical cells and circuits. We propose that there should be continued efforts to study cortical tissue from epilepsy patients in the quest to understand what makes human cell-types "human".
Collapse
Affiliation(s)
- Homeira Moradi Chameh
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
| | - Madeleine Falby
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Mandana Movahed
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
| | - Keon Arbabi
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Scott Rich
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Liang Zhang
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
| | - Jérémie Lefebvre
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Mathematics, University of Toronto, Toronto, ON, Canada
| | - Shreejoy J. Tripathy
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Maurizio De Pittà
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Basque Center for Applied Mathematics, Bilbao, Spain
- Faculty of Medicine, University of the Basque Country, Leioa, Spain
| | - Taufik A. Valiante
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network (UHN), Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, Canada
- Max Planck-University of Toronto Center for Neural Science and Technology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
de Kock CPJ, Feldmeyer D. Shared and divergent principles of synaptic transmission between cortical excitatory neurons in rodent and human brain. Front Synaptic Neurosci 2023; 15:1274383. [PMID: 37731775 PMCID: PMC10508294 DOI: 10.3389/fnsyn.2023.1274383] [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: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Information transfer between principal neurons in neocortex occurs through (glutamatergic) synaptic transmission. In this focussed review, we provide a detailed overview on the strength of synaptic neurotransmission between pairs of excitatory neurons in human and laboratory animals with a specific focus on data obtained using patch clamp electrophysiology. We reach two major conclusions: (1) the synaptic strength, measured as unitary excitatory postsynaptic potential (or uEPSP), is remarkably consistent across species, cortical regions, layers and/or cell-types (median 0.5 mV, interquartile range 0.4-1.0 mV) with most variability associated with the cell-type specific connection studied (min 0.1-max 1.4 mV), (2) synaptic function cannot be generalized across human and rodent, which we exemplify by discussing the differences in anatomical and functional properties of pyramidal-to-pyramidal connections within human and rodent cortical layers 2 and 3. With only a handful of studies available on synaptic transmission in human, it is obvious that much remains unknown to date. Uncovering the shared and divergent principles of synaptic transmission across species however, will almost certainly be a pivotal step toward understanding human cognitive ability and brain function in health and disease.
Collapse
Affiliation(s)
- Christiaan P. J. de Kock
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Dirk Feldmeyer
- Research Center Juelich, Institute of Neuroscience and Medicine, Jülich, Germany
- Department of Psychiatry, Psychotherapy, and Psychosomatics, RWTH Aachen University Hospital, Aachen, Germany
- Jülich-Aachen Research Alliance, Translational Brain Medicine (JARA Brain), Aachen, Germany
| |
Collapse
|
33
|
Inibhunu H, Moradi Chameh H, Skinner F, Rich S, Valiante TA. Hyperpolarization-Activated Cation Channels Shape the Spiking Frequency Preference of Human Cortical Layer 5 Pyramidal Neurons. eNeuro 2023; 10:ENEURO.0215-23.2023. [PMID: 37567768 PMCID: PMC10467019 DOI: 10.1523/eneuro.0215-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Discerning the contribution of specific ionic currents to complex neuronal dynamics is a difficult, but important, task. This challenge is exacerbated in the human setting, although the widely characterized uniqueness of the human brain compared with preclinical models necessitates the direct study of human neurons. Neuronal spiking frequency preference is of particular interest given its role in rhythm generation and signal transmission in cortical circuits. Here, we combine the frequency-dependent gain (FDG), a measure of spiking frequency preference, and novel in silico analyses to dissect the contributions of individual ionic currents to the suprathreshold features of human layer 5 (L5) neurons captured by the FDG. We confirm that a contemporary model of such a neuron, primarily constrained to capture subthreshold activity driven by the hyperpolarization-activated cyclic nucleotide gated (h-) current, replicates key features of the in vitro FDG both with and without h-current activity. With the model confirmed as a viable approximation of the biophysical features of interest, we applied new analysis techniques to quantify the activity of each modeled ionic current in the moments before spiking, revealing unique dynamics of the h-current. These findings motivated patch-clamp recordings in analogous rodent neurons to characterize their FDG, which confirmed that a biophysically detailed model of these neurons captures key interspecies differences in the FDG. These differences are correlated with distinct contributions of the h-current to neuronal activity. Together, this interdisciplinary and multispecies study provides new insights directly relating the dynamics of the h-current to suprathreshold spiking frequency preference in human L5 neurons.
Collapse
Affiliation(s)
- Happy Inibhunu
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 1M8, Canada
| | - Homeira Moradi Chameh
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 1M8, Canada
| | - Frances Skinner
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 1M8, Canada
- Departments of Medicine, Neurology and Physiology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Scott Rich
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 1M8, Canada
| | - Taufik A Valiante
- Division of Clinical and Computational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 1M8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3E2, Canada
- Electrical and Computer Engineering, University of Toronto, Toronto, Ontario M5S 3G4, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario M5T 1P5, Canada
| |
Collapse
|
34
|
Liu L, Yun Z, Manubens-Gil L, Chen H, Xiong F, Dong H, Zeng H, Hawrylycz M, Ascoli GA, Peng H. Neuronal Connectivity as a Determinant of Cell Types and Subtypes. RESEARCH SQUARE 2023:rs.3.rs-2960606. [PMID: 37398060 PMCID: PMC10312949 DOI: 10.21203/rs.3.rs-2960606/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Classifications of single neurons at brain-wide scale is a powerful way to characterize the structural and functional organization of a brain. We acquired and standardized a large morphology database of 20,158 mouse neurons, and generated a whole-brain scale potential connectivity map of single neurons based on their dendritic and axonal arbors. With such an anatomy-morphology-connectivity mapping, we defined neuron connectivity types and subtypes (both called "c-types" for simplicity) for neurons in 31 brain regions. We found that neuronal subtypes defined by connectivity in the same regions may share statistically higher correlation in their dendritic and axonal features than neurons having contrary connectivity patterns. Subtypes defined by connectivity show distinct separation with each other, which cannot be recapitulated by morphology features, population projections, transcriptomic, and electrophysiological data produced to date. Within this paradigm, we were able to characterize the diversity in secondary motor cortical neurons, and subtype connectivity patterns in thalamocortical pathways. Our finding underscores the importance of connectivity in characterizing the modularity of brain anatomy, as well as the cell types and their subtypes. These results highlight that c-types supplement conventionally recognized transcriptional cell types (t-types), electrophysiological cell types (e-types), and morphological cell types (m-types) as an important determinant of cell classes and their identities.
Collapse
Affiliation(s)
- Lijuan Liu
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu, China
| | - Zhixi Yun
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu, China
| | - Linus Manubens-Gil
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu, China
| | | | - Feng Xiong
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu, China
| | - Hongwei Dong
- UCLA Brain Research and Artificial Intelligence Nexus, Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Giorgio A. Ascoli
- Center for Neural Informatics, Bioengineering Department, and Neuroscience Program, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - Hanchuan Peng
- SEU-ALLEN Joint Center, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
35
|
Hunt S, Leibner Y, Mertens EJ, Barros-Zulaica N, Kanari L, Heistek TS, Karnani MM, Aardse R, Wilbers R, Heyer DB, Goriounova NA, Verhoog MB, Testa-Silva G, Obermayer J, Versluis T, Benavides-Piccione R, de Witt-Hamer P, Idema S, Noske DP, Baayen JC, Lein ES, DeFelipe J, Markram H, Mansvelder HD, Schürmann F, Segev I, de Kock CPJ. Strong and reliable synaptic communication between pyramidal neurons in adult human cerebral cortex. Cereb Cortex 2023; 33:2857-2878. [PMID: 35802476 PMCID: PMC10016070 DOI: 10.1093/cercor/bhac246] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/25/2022] Open
Abstract
Synaptic transmission constitutes the primary mode of communication between neurons. It is extensively studied in rodent but not human neocortex. We characterized synaptic transmission between pyramidal neurons in layers 2 and 3 using neurosurgically resected human middle temporal gyrus (MTG, Brodmann area 21), which is part of the distributed language circuitry. We find that local connectivity is comparable with mouse layer 2/3 connections in the anatomical homologue (temporal association area), but synaptic connections in human are 3-fold stronger and more reliable (0% vs 25% failure rates, respectively). We developed a theoretical approach to quantify properties of spinous synapses showing that synaptic conductance and voltage change in human dendritic spines are 3-4-folds larger compared with mouse, leading to significant NMDA receptor activation in human unitary connections. This model prediction was validated experimentally by showing that NMDA receptor activation increases the amplitude and prolongs decay of unitary excitatory postsynaptic potentials in human but not in mouse connections. Since NMDA-dependent recurrent excitation facilitates persistent activity (supporting working memory), our data uncovers cortical microcircuit properties in human that may contribute to language processing in MTG.
Collapse
Affiliation(s)
| | | | - Eline J Mertens
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Natalí Barros-Zulaica
- Blue Brain Project, Ecole polytechnique fédérale de Lausanne, Campus Biotech, Geneva 1202, Switzerland
| | - Lida Kanari
- Blue Brain Project, Ecole polytechnique fédérale de Lausanne, Campus Biotech, Geneva 1202, Switzerland
| | - Tim S Heistek
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Mahesh M Karnani
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Romy Aardse
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - René Wilbers
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Djai B Heyer
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Natalia A Goriounova
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | | | | | - Joshua Obermayer
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Tamara Versluis
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Ruth Benavides-Piccione
- Laboratorio Cajal de Circuitos Corticales, Universidad Politécnica de Madrid and Instituto Cajal (CSIC), Pozuelo de Alarcón, Madrid 28223, Spain
| | - Philip de Witt-Hamer
- Neurosurgery Department, Amsterdam Universitair Medische Centra, location VUmc, 1081 HV Amsterdam, the Netherlands
| | - Sander Idema
- Neurosurgery Department, Amsterdam Universitair Medische Centra, location VUmc, 1081 HV Amsterdam, the Netherlands
| | - David P Noske
- Neurosurgery Department, Amsterdam Universitair Medische Centra, location VUmc, 1081 HV Amsterdam, the Netherlands
| | - Johannes C Baayen
- Neurosurgery Department, Amsterdam Universitair Medische Centra, location VUmc, 1081 HV Amsterdam, the Netherlands
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Universidad Politécnica de Madrid and Instituto Cajal (CSIC), Pozuelo de Alarcón, Madrid 28223, Spain
| | - Henry Markram
- Blue Brain Project, Ecole polytechnique fédérale de Lausanne, Campus Biotech, Geneva 1202, Switzerland
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Felix Schürmann
- Blue Brain Project, Ecole polytechnique fédérale de Lausanne, Campus Biotech, Geneva 1202, Switzerland
| | - Idan Segev
- Department of Neurobiology and Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, 9190501 Jerusalem, Israel
| | | |
Collapse
|
36
|
Vanderhaeghen P, Polleux F. Developmental mechanisms underlying the evolution of human cortical circuits. Nat Rev Neurosci 2023; 24:213-232. [PMID: 36792753 PMCID: PMC10064077 DOI: 10.1038/s41583-023-00675-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 02/17/2023]
Abstract
The brain of modern humans has evolved remarkable computational abilities that enable higher cognitive functions. These capacities are tightly linked to an increase in the size and connectivity of the cerebral cortex, which is thought to have resulted from evolutionary changes in the mechanisms of cortical development. Convergent progress in evolutionary genomics, developmental biology and neuroscience has recently enabled the identification of genomic changes that act as human-specific modifiers of cortical development. These modifiers influence most aspects of corticogenesis, from the timing and complexity of cortical neurogenesis to synaptogenesis and the assembly of cortical circuits. Mutations of human-specific genetic modifiers of corticogenesis have started to be linked to neurodevelopmental disorders, providing evidence for their physiological relevance and suggesting potential relationships between the evolution of the human brain and its sensitivity to specific diseases.
Collapse
Affiliation(s)
- Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| | - Franck Polleux
- Department of Neuroscience, Columbia University Medical Center, New York, NY, USA.
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
| |
Collapse
|
37
|
Renner J, Rasia-Filho AA. Morphological Features of Human Dendritic Spines. ADVANCES IN NEUROBIOLOGY 2023; 34:367-496. [PMID: 37962801 DOI: 10.1007/978-3-031-36159-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dendritic spine features in human neurons follow the up-to-date knowledge presented in the previous chapters of this book. Human dendrites are notable for their heterogeneity in branching patterns and spatial distribution. These data relate to circuits and specialized functions. Spines enhance neuronal connectivity, modulate and integrate synaptic inputs, and provide additional plastic functions to microcircuits and large-scale networks. Spines present a continuum of shapes and sizes, whose number and distribution along the dendritic length are diverse in neurons and different areas. Indeed, human neurons vary from aspiny or "relatively aspiny" cells to neurons covered with a high density of intermingled pleomorphic spines on very long dendrites. In this chapter, we discuss the phylogenetic and ontogenetic development of human spines and describe the heterogeneous features of human spiny neurons along the spinal cord, brainstem, cerebellum, thalamus, basal ganglia, amygdala, hippocampal regions, and neocortical areas. Three-dimensional reconstructions of Golgi-impregnated dendritic spines and data from fluorescence microscopy are reviewed with ultrastructural findings to address the complex possibilities for synaptic processing and integration in humans. Pathological changes are also presented, for example, in Alzheimer's disease and schizophrenia. Basic morphological data can be linked to current techniques, and perspectives in this research field include the characterization of spines in human neurons with specific transcriptome features, molecular classification of cellular diversity, and electrophysiological identification of coexisting subpopulations of cells. These data would enlighten how cellular attributes determine neuron type-specific connectivity and brain wiring for our diverse aptitudes and behavior.
Collapse
Affiliation(s)
- Josué Renner
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Alberto A Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
38
|
Testa-Silva G, Rosier M, Honnuraiah S, Guzulaitis R, Megias AM, French C, King J, Drummond K, Palmer LM, Stuart GJ. High synaptic threshold for dendritic NMDA spike generation in human layer 2/3 pyramidal neurons. Cell Rep 2022; 41:111787. [PMID: 36516769 DOI: 10.1016/j.celrep.2022.111787] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/19/2022] [Accepted: 11/15/2022] [Indexed: 12/15/2022] Open
Abstract
Neurons receive synaptic input primarily onto their dendrites. While we know much about the electrical properties of dendrites in rodents, we have only just started to describe their properties in the human brain. Here, we investigate the capacity of human dendrites to generate NMDA-receptor-dependent spikes (NMDA spikes). Using dendritic glutamate iontophoresis, as well as local dendritic synaptic stimulation, we find that human layer 2/3 pyramidal neurons can generate dendritic NMDA spikes. The capacity to evoke NMDA spikes in human neurons, however, was significantly reduced compared with that in rodents. Simulations in morphologically realistic and simplified models indicated that human neurons have a higher synaptic threshold for NMDA spike generation primarily due to the wider diameter of their dendrites. In summary, we find reduced NMDA spike generation in human compared with rodent layer 2/3 pyramidal neurons and provide evidence that this is due to the wider diameter of human dendrites.
Collapse
Affiliation(s)
- Guilherme Testa-Silva
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Marius Rosier
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia; Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Suraj Honnuraiah
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Robertas Guzulaitis
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Ana Morello Megias
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Chris French
- The University of Melbourne, Department of Surgery, Parkville, VIC, Australia; The Royal Melbourne Hospital, Department of Neurosurgery, Parkville, VIC, Australia
| | - James King
- The Royal Melbourne Hospital, Department of Neurosurgery, Parkville, VIC, Australia
| | - Katharine Drummond
- The University of Melbourne, Department of Surgery, Parkville, VIC, Australia; The Royal Melbourne Hospital, Department of Neurosurgery, Parkville, VIC, Australia
| | - Lucy M Palmer
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia.
| | - Greg J Stuart
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia; Department of Physiology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
39
|
Howard D, Chameh HM, Guet-McCreight A, Hsiao HA, Vuong M, Seo YS, Shah P, Nigam A, Chen Y, Davie M, Hay E, Valiante TA, Tripathy SJ. An in vitro whole-cell electrophysiology dataset of human cortical neurons. Gigascience 2022; 11:giac108. [PMID: 36377463 PMCID: PMC9664072 DOI: 10.1093/gigascience/giac108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/11/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Whole-cell patch-clamp electrophysiology is an essential technique for understanding how single neurons translate their diverse inputs into a functional output. The relative inaccessibility of live human cortical neurons for experimental manipulation has made it difficult to determine the unique features of how human cortical neurons differ from their counterparts in other species. FINDINGS We present a curated repository of whole-cell patch-clamp recordings from surgically resected human cortical tissue, encompassing 118 neurons from 35 individuals (age range, 21-59 years; 17 male, 18 female). Recorded human cortical neurons derive from layers 2 and 3 (L2&3), deep layer 3 (L3c), or layer 5 (L5) and are annotated with a rich set of subject and experimental metadata. For comparison, we also provide a limited set of comparable recordings from 21-day-old mice (11 cells from 5 mice). All electrophysiological recordings are provided in the Neurodata Without Borders (NWB) format and are available for further analysis via the Distributed Archives for Neurophysiology Data Integration online repository. The associated data conversion code is made publicly available and can help others in converting electrophysiology datasets to the open NWB standard for general reuse. CONCLUSION These data can be used for novel analyses of biophysical characteristics of human cortical neurons, including in cross-species or cross-lab comparisons or in building computational models of individual human neurons.
Collapse
Affiliation(s)
- Derek Howard
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
| | | | - Alexandre Guet-McCreight
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
| | - Huan Allen Hsiao
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
| | - Maggie Vuong
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
| | - Young Seok Seo
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
| | - Prajay Shah
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
| | - Anukrati Nigam
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Yuxiao Chen
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
| | - Melanie Davie
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
| | - Etay Hay
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Taufik A Valiante
- Krembil Brain Institute, University Health Network, Toronto, ON, M5T 1M8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON, M5S 1A4, Canada
- Department of Surgery, Division of Neurosurgery, University of Toronto, Toronto, ON, M5T 1P5, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
- Max Planck–University of Toronto Center for Neural Science and Technology, Toronto, ON, M5S 1A4, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, ON , M5S 1A4, Canada
| | - Shreejoy J Tripathy
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Surgery, Division of Neurosurgery, University of Toronto, Toronto, ON, M5T 1P5, Canada
- Department of Psychiatry, University of Toront, Toronto, ON, M5T 1R8, Canada
| |
Collapse
|
40
|
Guerra KTK, Renner J, Vásquez CE, Rasia‐Filho AA. Human cortical amygdala dendrites and spines morphology under open‐source three‐dimensional reconstruction procedures. J Comp Neurol 2022; 531:344-365. [PMID: 36355397 DOI: 10.1002/cne.25430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/05/2022] [Accepted: 10/14/2022] [Indexed: 11/12/2022]
Abstract
Visualizing nerve cells has been fundamental for the systematic description of brain structure and function in humans and other species. Different approaches aimed to unravel the morphological features of neuron types and diversity. The inherent complexity of the human nervous tissue and the need for proper histological processing have made studying human dendrites and spines challenging in postmortem samples. In this study, we used Golgi data and open-source software for 3D image reconstruction of human neurons from the cortical amygdaloid nucleus to show different dendrites and pleomorphic spines at different angles. Procedures required minimal equipment and generated high-quality images for differently shaped cells. We used the "single-section" Golgi method adapted for the human brain to engender 3D reconstructed images of the neuronal cell body and the dendritic ramification by adopting a neuronal tracing procedure. In addition, we elaborated 3D reconstructions to visualize heterogeneous dendritic spines using a supervised machine learning-based algorithm for image segmentation. These tools provided an additional upgrade and enhanced visual display of information related to the spatial orientation of dendritic branches and for dendritic spines of varied sizes and shapes in these human subcortical neurons. This same approach can be adapted for other techniques, areas of the central or peripheral nervous system, and comparative analysis between species.
Collapse
Affiliation(s)
- Kétlyn T. Knak Guerra
- Graduate Program in Neuroscience Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Josué Renner
- Department of Basic Sciences/Physiology Universidade Federal de Ciências da Saúde de Porto Alegre Porto Alegre Brazil
- Graduate Program in Biosciences Universidade Federal de Ciências da Saúde de Porto Alegre Porto Alegre Brazil
| | - Carlos E. Vásquez
- Graduate Program in Neuroscience Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Alberto A. Rasia‐Filho
- Graduate Program in Neuroscience Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
- Department of Basic Sciences/Physiology Universidade Federal de Ciências da Saúde de Porto Alegre Porto Alegre Brazil
- Graduate Program in Biosciences Universidade Federal de Ciências da Saúde de Porto Alegre Porto Alegre Brazil
| |
Collapse
|
41
|
Galakhova AA, Hunt S, Wilbers R, Heyer DB, de Kock CPJ, Mansvelder HD, Goriounova NA. Evolution of cortical neurons supporting human cognition. Trends Cogn Sci 2022; 26:909-922. [PMID: 36117080 PMCID: PMC9561064 DOI: 10.1016/j.tics.2022.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 01/12/2023]
Abstract
Human cognitive abilities are generally thought to arise from cortical expansion over the course of human brain evolution. In addition to increased neuron numbers, this cortical expansion might be driven by adaptations in the properties of single neurons and their local circuits. We review recent findings on the distinct structural, functional, and transcriptomic features of human cortical neurons and their organization in cortical microstructure. We focus on the supragranular cortical layers, which showed the most prominent expansion during human brain evolution, and the properties of their principal cells: pyramidal neurons. We argue that the evolutionary adaptations in neuronal features that accompany the expansion of the human cortex partially underlie interindividual variability in human cognitive abilities.
Collapse
Affiliation(s)
- A A Galakhova
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - S Hunt
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - R Wilbers
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - D B Heyer
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - C P J de Kock
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - H D Mansvelder
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands
| | - N A Goriounova
- Department of Integrative Neurophysiology, Amsterdam Neuroscience, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, The Netherlands.
| |
Collapse
|
42
|
Moberg S, Takahashi N. Neocortical layer 5 subclasses: From cellular properties to roles in behavior. Front Synaptic Neurosci 2022; 14:1006773. [PMID: 36387773 PMCID: PMC9650089 DOI: 10.3389/fnsyn.2022.1006773] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/28/2022] [Indexed: 09/08/2024] Open
Abstract
Layer 5 (L5) serves as the main output layer of cortical structures, where long-range projecting pyramidal neurons broadcast the columnar output to other cortical and extracortical regions of the brain. L5 pyramidal neurons are grouped into two subclasses based on their projection targets; while intratelencephalic (IT) neurons project to cortical areas and the striatum, extratelencephalic (ET) neurons project to subcortical areas such as the thalamus, midbrain, and brainstem. Each L5 subclass possesses distinct morphological and electrophysiological properties and is incorporated into a unique synaptic network. Thanks to recent advances in genetic tools and methodologies, it has now become possible to distinguish between the two subclasses in the living brain. There is increasing evidence indicating that each subclass plays a unique role in sensory processing, decision-making, and learning. This review first summarizes the anatomical and physiological properties as well as the neuromodulation of IT and ET neurons in the rodent neocortex, and then reviews recent literature on their roles in sensory processing and rodent behavior. Our ultimate goal is to provide a comprehensive understanding of the role of each subclass in cortical function by examining their operational regimes based on their cellular properties.
Collapse
Affiliation(s)
- Sara Moberg
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Naoya Takahashi
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| |
Collapse
|
43
|
Moore H, Lega BC, Konopka G. Riding brain "waves" to identify human memory genes. Curr Opin Cell Biol 2022; 78:102118. [PMID: 35947942 DOI: 10.1016/j.ceb.2022.102118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 01/31/2023]
Abstract
While there is extensive research on memory-related oscillations and brain gene expression, the relationship between oscillations and gene expression has rarely been studied. Recently, progress has been made to identify specific genes associated with oscillations that are correlated with episodic memory. Neocortical regions, in particular the temporal pole, have been examined in this line of research due to their accessibility during neurosurgical procedures. By harnessing this accessibility, a unique and powerful study design has allowed gene expression and intracranial oscillatory data to be sourced from the same human patients. These studies have identified a plethora of understudied gene targets that should be further characterized with respect to human brain function. Future work should extend to other brain regions to increase our understanding of the genetic signatures of oscillations and, ultimately, human cognition.
Collapse
Affiliation(s)
- Haley Moore
- Department of Neuroscience, UT Southwestern Medical Center, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, USA; Department of Neurosurgery, UT Southwestern Medical Center, USA
| | - Bradley C Lega
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, USA; Department of Neurosurgery, UT Southwestern Medical Center, USA.
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, USA.
| |
Collapse
|
44
|
Gooch HM, Bluett T, Perumal MB, Vo HD, Fletcher LN, Papacostas J, Jeffree RL, Wood M, Colditz MJ, McMillen J, Tsahtsarlis T, Amato D, Campbell R, Gillinder L, Williams SR. High-fidelity dendritic sodium spike generation in human layer 2/3 neocortical pyramidal neurons. Cell Rep 2022; 41:111500. [DOI: 10.1016/j.celrep.2022.111500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/22/2022] [Accepted: 09/21/2022] [Indexed: 11/03/2022] Open
|
45
|
Guet-McCreight A, Chameh HM, Mahallati S, Wishart M, Tripathy SJ, Valiante TA, Hay E. Age-dependent increased sag amplitude in human pyramidal neurons dampens baseline cortical activity. Cereb Cortex 2022; 33:4360-4373. [PMID: 36124673 DOI: 10.1093/cercor/bhac348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 11/14/2022] Open
Abstract
Aging involves various neurobiological changes, although their effect on brain function in humans remains poorly understood. The growing availability of human neuronal and circuit data provides opportunities for uncovering age-dependent changes of brain networks and for constraining models to predict consequences on brain activity. Here we found increased sag voltage amplitude in human middle temporal gyrus layer 5 pyramidal neurons from older subjects and captured this effect in biophysical models of younger and older pyramidal neurons. We used these models to simulate detailed layer 5 microcircuits and found lower baseline firing in older pyramidal neuron microcircuits, with minimal effect on response. We then validated the predicted reduced baseline firing using extracellular multielectrode recordings from human brain slices of different ages. Our results thus report changes in human pyramidal neuron input integration properties and provide fundamental insights into the neuronal mechanisms of altered cortical excitability and resting-state activity in human aging.
Collapse
Affiliation(s)
- Alexandre Guet-McCreight
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, 250 College St, Toronto, ON M5T 1R8, Canada
| | | | - Sara Mahallati
- Krembil Brain Institute, University Health Network, Toronto, ON M5T1M8, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Margaret Wishart
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, 250 College St, Toronto, ON M5T 1R8, Canada
| | - Shreejoy J Tripathy
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, 250 College St, Toronto, ON M5T 1R8, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada.,Department of Physiology, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Taufik A Valiante
- Krembil Brain Institute, University Health Network, Toronto, ON M5T1M8, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada.,Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S 3G4, Canada.,Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada.,Center for Advancing Neurotechnological Innovation to Application, University of Toronto, Toronto, ON M5G 2A2, Canada.,Max Planck-University of Toronto Center for Neural Science and Technology, Toronto, ON, Canada
| | - Etay Hay
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, 250 College St, Toronto, ON M5T 1R8, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada.,Department of Physiology, University of Toronto, Toronto, ON M5S1A8, Canada
| |
Collapse
|
46
|
Pavón Arocas O, Branco T. Preparation of acute midbrain slices containing the superior colliculus and periaqueductal Gray for patch-clamp recordings. PLoS One 2022; 17:e0271832. [PMID: 35951507 PMCID: PMC9371254 DOI: 10.1371/journal.pone.0271832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/07/2022] [Indexed: 11/18/2022] Open
Abstract
This protocol is a practical guide for preparing acute coronal slices from the midbrain of young adult mice for electrophysiology experiments. It describes two different sets of solutions with their respective incubation strategies and two alternative procedures for brain extraction: decapitation under terminal isoflurane anaesthesia and intracardial perfusion with artificial cerebrospinal fluid under terminal isoflurane anaesthesia. Slices can be prepared from wild-type mice as well as from mice that have been genetically modified or transfected with viral constructs to label subsets of cells. The preparation can be used to investigate the electrophysiological properties of midbrain neurons in combination with pharmacology, opto- and chemogenetic manipulations, and calcium imaging; which can be followed by morphological reconstruction, immunohistochemistry, or single-cell transcriptomics. The protocol also provides a detailed list of materials and reagents including the design for a low-cost and easy to assemble 3D printed slice recovery chamber, general advice for troubleshooting common issues leading to suboptimal slice quality, and some suggestions to ensure good maintenance of a patch-clamp rig.
Collapse
Affiliation(s)
- Oriol Pavón Arocas
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, United Kingdom
| | - Tiago Branco
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, United Kingdom
| |
Collapse
|
47
|
Jedlicka P, Bird AD, Cuntz H. Pareto optimality, economy-effectiveness trade-offs and ion channel degeneracy: improving population modelling for single neurons. Open Biol 2022; 12:220073. [PMID: 35857898 PMCID: PMC9277232 DOI: 10.1098/rsob.220073] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Neurons encounter unavoidable evolutionary trade-offs between multiple tasks. They must consume as little energy as possible while effectively fulfilling their functions. Cells displaying the best performance for such multi-task trade-offs are said to be Pareto optimal, with their ion channel configurations underpinning their functionality. Ion channel degeneracy, however, implies that multiple ion channel configurations can lead to functionally similar behaviour. Therefore, instead of a single model, neuroscientists often use populations of models with distinct combinations of ionic conductances. This approach is called population (database or ensemble) modelling. It remains unclear, which ion channel parameters in the vast population of functional models are more likely to be found in the brain. Here we argue that Pareto optimality can serve as a guiding principle for addressing this issue by helping to identify the subpopulations of conductance-based models that perform best for the trade-off between economy and functionality. In this way, the high-dimensional parameter space of neuronal models might be reduced to geometrically simple low-dimensional manifolds, potentially explaining experimentally observed ion channel correlations. Conversely, Pareto inference might also help deduce neuronal functions from high-dimensional Patch-seq data. In summary, Pareto optimality is a promising framework for improving population modelling of neurons and their circuits.
Collapse
Affiliation(s)
- Peter Jedlicka
- ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany,Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt/Main, Germany,Frankfurt Institute for Advanced Studies, Frankfurt am Main, Germany
| | - Alexander D. Bird
- ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany,Frankfurt Institute for Advanced Studies, Frankfurt am Main, Germany,Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Frankfurt am Main, Germany
| | - Hermann Cuntz
- Frankfurt Institute for Advanced Studies, Frankfurt am Main, Germany,Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Frankfurt am Main, Germany
| |
Collapse
|
48
|
Loss of neuronal heterogeneity in epileptogenic human tissue impairs network resilience to sudden changes in synchrony. Cell Rep 2022; 39:110863. [PMID: 35613586 DOI: 10.1016/j.celrep.2022.110863] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 03/16/2022] [Accepted: 05/03/2022] [Indexed: 12/25/2022] Open
Abstract
A myriad of pathological changes associated with epilepsy can be recast as decreases in cell and circuit heterogeneity. We thus propose recontextualizing epileptogenesis as a process where reduction in cellular heterogeneity, in part, renders neural circuits less resilient to seizure. By comparing patch clamp recordings from human layer 5 (L5) cortical pyramidal neurons from epileptogenic and non-epileptogenic tissue, we demonstrate significantly decreased biophysical heterogeneity in seizure-generating areas. Implemented computationally, this renders model neural circuits prone to sudden transitions into synchronous states with increased firing activity, paralleling ictogenesis. This computational work also explains the surprising finding of significantly decreased excitability in the population-activation functions of neurons from epileptogenic tissue. Finally, mathematical analyses reveal a bifurcation structure arising only with low heterogeneity and associated with seizure-like dynamics. Taken together, this work provides experimental, computational, and mathematical support for the theory that ictogenic dynamics accompany a reduction in biophysical heterogeneity.
Collapse
|
49
|
Tian D, Izumi SI. Transcranial Magnetic Stimulation and Neocortical Neurons: The Micro-Macro Connection. Front Neurosci 2022; 16:866245. [PMID: 35495053 PMCID: PMC9039343 DOI: 10.3389/fnins.2022.866245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/28/2022] [Indexed: 12/20/2022] Open
Abstract
Understanding the operation of cortical circuits is an important and necessary task in both neuroscience and neurorehabilitation. The functioning of the neocortex results from integrative neuronal activity, which can be probed non-invasively by transcranial magnetic stimulation (TMS). Despite a clear indication of the direct involvement of cortical neurons in TMS, no explicit connection model has been made between the microscopic neuronal landscape and the macroscopic TMS outcome. Here we have performed an integrative review of multidisciplinary evidence regarding motor cortex neurocytology and TMS-related neurophysiology with the aim of elucidating the micro–macro connections underlying TMS. Neurocytological evidence from animal and human studies has been reviewed to describe the landscape of the cortical neurons covering the taxonomy, morphology, circuit wiring, and excitatory–inhibitory balance. Evidence from TMS studies in healthy humans is discussed, with emphasis on the TMS pulse and paradigm selectivity that reflect the underlying neural circuitry constitution. As a result, we propose a preliminary neuronal model of the human motor cortex and then link the TMS mechanisms with the neuronal model by stimulus intensity, direction of induced current, and paired-pulse timing. As TMS bears great developmental potential for both a probe and modulator of neural network activity and neurotransmission, the connection model will act as a foundation for future combined studies of neurocytology and neurophysiology, as well as the technical advances and application of TMS.
Collapse
Affiliation(s)
- Dongting Tian
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduates School of Medicine, Sendai, Japan
- *Correspondence: Dongting Tian,
| | - Shin-Ichi Izumi
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduates School of Medicine, Sendai, Japan
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
- Shin-Ichi Izumi,
| |
Collapse
|
50
|
Rapti G. Open Frontiers in Neural Cell Type Investigations; Lessons From Caenorhabditis elegans and Beyond, Toward a Multimodal Integration. Front Neurosci 2022; 15:787753. [PMID: 35321480 PMCID: PMC8934944 DOI: 10.3389/fnins.2021.787753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Nervous system cells, the building blocks of circuits, have been studied with ever-progressing resolution, yet neural circuits appear still resistant to schemes of reductionist classification. Due to their sheer numbers, complexity and diversity, their systematic study requires concrete classifications that can serve reduced dimensionality, reproducibility, and information integration. Conventional hierarchical schemes transformed through the history of neuroscience by prioritizing criteria of morphology, (electro)physiological activity, molecular content, and circuit function, influenced by prevailing methodologies of the time. Since the molecular biology revolution and the recent advents in transcriptomics, molecular profiling gains ground toward the classification of neurons and glial cell types. Yet, transcriptomics entails technical challenges and more importantly uncovers unforeseen spatiotemporal heterogeneity, in complex and simpler nervous systems. Cells change states dynamically in space and time, in response to stimuli or throughout their developmental trajectory. Mapping cell type and state heterogeneity uncovers uncharted terrains in neurons and especially in glial cell biology, that remains understudied in many aspects. Examining neurons and glial cells from the perspectives of molecular neuroscience, physiology, development and evolution highlights the advantage of multifaceted classification schemes. Among the amalgam of models contributing to neuroscience research, Caenorhabditis elegans combines nervous system anatomy, lineage, connectivity and molecular content, all mapped at single-cell resolution, and can provide valuable insights for the workflow and challenges of the multimodal integration of cell type features. This review reflects on concepts and practices of neuron and glial cells classification and how research, in C. elegans and beyond, guides nervous system experimentation through integrated multidimensional schemes. It highlights underlying principles, emerging themes, and open frontiers in the study of nervous system development, regulatory logic and evolution. It proposes unified platforms to allow integrated annotation of large-scale datasets, gene-function studies, published or unpublished findings and community feedback. Neuroscience is moving fast toward interdisciplinary, high-throughput approaches for combined mapping of the morphology, physiology, connectivity, molecular function, and the integration of information in multifaceted schemes. A closer look in mapped neural circuits and understudied terrains offers insights for the best implementation of these approaches.
Collapse
|