1
|
Chung EN, Lee J, Polonio CM, Choi J, Akl CF, Kilian M, Weiß WM, Gunner G, Ye M, Heo TH, Drake SS, Yang L, d'Eca CRGL, Lee JH, Deng L, Farrenkopf D, Schüle AM, Lee HG, Afolabi O, Ghaznavi S, Smirnakis SM, Chiu IM, Kuchroo VK, Quintana FJ, Wheeler MA. Psychedelic control of neuroimmune interactions governing fear. Nature 2025; 641:1276-1286. [PMID: 40269152 PMCID: PMC12119215 DOI: 10.1038/s41586-025-08880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 03/11/2025] [Indexed: 04/25/2025]
Abstract
Neuroimmune interactions-signals transmitted between immune and brain cells-regulate many aspects of tissue physiology1, including responses to psychological stress2-5, which can predispose individuals to develop neuropsychiatric diseases6-9. Still, the interactions between haematopoietic and brain-resident cells that influence complex behaviours are poorly understood. Here, we use a combination of genomic and behavioural screens to show that astrocytes in the amygdala limit stress-induced fear behaviour through epidermal growth factor receptor (EGFR). Mechanistically, EGFR expression in amygdala astrocytes inhibits a stress-induced, pro-inflammatory signal-transduction cascade that facilitates neuron-glial crosstalk and stress-induced fear behaviour through the orphan nuclear receptor NR2F2 in amygdala neurons. In turn, decreased EGFR signalling and fear behaviour are associated with the recruitment of meningeal monocytes during chronic stress. This set of neuroimmune interactions is therapeutically targetable through the administration of psychedelic compounds, which reversed the accumulation of monocytes in the brain meninges along with fear behaviour. Together with validation in clinical samples, these data suggest that psychedelics can be used to target neuroimmune interactions relevant to neuropsychiatric disorders and potentially other inflammatory diseases.
Collapse
Affiliation(s)
- Elizabeth N Chung
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Jinsu Lee
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Carolina M Polonio
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Joshua Choi
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Camilo Faust Akl
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael Kilian
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Wiebke M Weiß
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Georgia Gunner
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Mingyu Ye
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA, USA
| | - Tae Hyun Heo
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Sienna S Drake
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Liu Yang
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Catarina R G L d'Eca
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Joon-Hyuk Lee
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Liwen Deng
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Daniel Farrenkopf
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Anton M Schüle
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Hong-Gyun Lee
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Oreoluwa Afolabi
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Sharmin Ghaznavi
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for the Neuroscience of Psychedelics, Massachusetts General Hospital, Boston, MA, USA
| | - Stelios M Smirnakis
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA, USA
| | - Isaac M Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Vijay K Kuchroo
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Francisco J Quintana
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael A Wheeler
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
2
|
Jansma A, Yao Y, Wolfe J, Del Debbio L, Beentjes SV, Ponting CP, Khamseh A. High order expression dependencies finely resolve cryptic states and subtypes in single cell data. Mol Syst Biol 2025; 21:173-207. [PMID: 39748128 PMCID: PMC11790937 DOI: 10.1038/s44320-024-00074-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 01/04/2025] Open
Abstract
Single cells are typically typed by clustering into discrete locations in reduced dimensional transcriptome space. Here we introduce Stator, a data-driven method that identifies cell (sub)types and states without relying on cells' local proximity in transcriptome space. Stator labels the same single cell multiply, not just by type and subtype, but also by state such as activation, maturity or cell cycle sub-phase, through deriving higher-order gene expression dependencies from a sparse gene-by-cell expression matrix. Stator's finer resolution is clear from analyses of mouse embryonic brain, and human healthy or diseased liver. Rather than only coarse-scale labels of cell type, Stator further resolves cell types into subtypes, and these subtypes into stages of maturity and/or cell cycle phases, and yet further into portions of these phases. Among cryptically homogeneous embryonic cells, for example, Stator finds 34 distinct radial glia states whose gene expression forecasts their future GABAergic or glutamatergic neuronal fate. Further, Stator's fine resolution of liver cancer states reveals expression programmes that predict patient survival. We provide Stator as a Nextflow pipeline and Shiny App.
Collapse
Affiliation(s)
- Abel Jansma
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
- Higgs Centre for Theoretical Physics, School of Physics & Astronomy, University of Edinburgh, Edinburgh, EH9 3FD, UK
| | - Yuelin Yao
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
- School of Informatics, University of Edinburgh, Edinburgh, EH8 9AB, UK
| | - Jareth Wolfe
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Luigi Del Debbio
- Higgs Centre for Theoretical Physics, School of Physics & Astronomy, University of Edinburgh, Edinburgh, EH9 3FD, UK
| | - Sjoerd V Beentjes
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
- School of Mathematics, University of Edinburgh, Edinburgh, EH9 3FD, UK
| | - Chris P Ponting
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK.
| | - Ava Khamseh
- MRC Human Genetics Unit, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK.
- Higgs Centre for Theoretical Physics, School of Physics & Astronomy, University of Edinburgh, Edinburgh, EH9 3FD, UK.
- School of Informatics, University of Edinburgh, Edinburgh, EH8 9AB, UK.
| |
Collapse
|
3
|
Tweedie L, Riccetti MR, Cain B, Qin S, Salomone J, Webb JA, Riesenberg A, Ehrman LA, Waclaw RR, Kovall RA, Gebelein B, Campbell K. Modelling a pathological GSX2 variant that selectively alters DNA binding reveals hypomorphic mouse brain defects. Dis Model Mech 2025; 18:dmm052110. [PMID: 39882631 PMCID: PMC11876842 DOI: 10.1242/dmm.052110] [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: 09/17/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
Gsx2 is a homeodomain transcription factor critical for development of the ventral telencephalon and hindbrain in mouse. Loss of Gsx2 function results in severe basal ganglia dysgenesis and defects in the nucleus tractus solitarius (nTS) of the hindbrain, together with respiratory failure at birth. De Mori et al. (2019) reported two patients with severe dystonia and basal ganglia dysgenesis that encode distinct recessive GSX2 variants, including a missense variant within the homeodomain (GSX2Q251R). Hence, we modelled the homologous Gsx2 mutation (i.e. Gsx2Q252R) in mouse, and our biochemical analysis revealed that this variant selectively altered DNA binding. Moreover, mice carrying the Gsx2Q252R allele exhibited basal ganglia dysgenesis, albeit to a lesser extent than did Gsx2 null mice. A notable difference between Gsx2Q252R and Gsx2 null mice was that Gsx2Q252R mice survived, and hindbrain analysis revealed relative sparing of the glutamatergic nTS neurons and catecholaminergic A1/C1 and A2/C2 groups. Thus, the Gsx2Q252R variant is a hypomorph that compromises a subset of Gsx2-dependent neuronal subtypes and highlights a critical role for distinct thresholds of catecholaminergic and/or glutamatergic nTS neurons for viability.
Collapse
Affiliation(s)
- Laura Tweedie
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Matthew R. Riccetti
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Brittany Cain
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Shenyue Qin
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Joseph Salomone
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jordan A. Webb
- Department of Molecular and Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Amy Riesenberg
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Lisa A. Ehrman
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Ronald R. Waclaw
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Rhett A. Kovall
- Department of Molecular and Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Brian Gebelein
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Kenneth Campbell
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
4
|
Aksoy-Aksel A, Ferraguti F, Holmes A, Lüthi A, Ehrlich I. Amygdala intercalated cells form an evolutionarily conserved system orchestrating brain networks. Nat Neurosci 2025; 28:234-247. [PMID: 39672964 DOI: 10.1038/s41593-024-01836-8] [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: 02/28/2024] [Accepted: 11/01/2024] [Indexed: 12/15/2024]
Abstract
The amygdala attributes valence and emotional salience to environmental stimuli and regulates how these stimuli affect behavior. Within the amygdala, a distinct class of evolutionarily conserved neurons form the intercalated cell (ITC) clusters, mainly located around the boundaries of the lateral and basal nuclei. Here, we review the anatomical, physiological and molecular characteristics of ITCs, and detail the organization of ITC clusters and their connectivity with one another and other brain regions. We describe how ITCs undergo experience-dependent plasticity and discuss emerging evidence demonstrating how ITCs are innervated and functionally regulated by neuromodulatory systems. We summarize recent findings showing that experience alters the balance of activity between different ITC clusters, thereby determining prevailing behavioral output. Finally, we propose a model in which ITCs form a key system for integrating divergent inputs and orchestrating brain-wide circuits to generate behavioral states attuned to current environmental circumstances and internal needs.
Collapse
Affiliation(s)
- Ayla Aksoy-Aksel
- Department of Neurobiology, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - Andreas Lüthi
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Ingrid Ehrlich
- Department of Neurobiology, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
5
|
Kamboj S, Carlson EL, Ander BP, Hanson KL, Murray KD, Fudge JL, Bauman MD, Schumann CM, Fox AS. Translational Insights From Cell Type Variation Across Amygdala Subnuclei in Rhesus Monkeys and Humans. Am J Psychiatry 2024; 181:1086-1102. [PMID: 39473267 DOI: 10.1176/appi.ajp.20230602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
OBJECTIVE Theories of amygdala function are central to our understanding of psychiatric and neurodevelopmental disorders. However, limited knowledge of the molecular and cellular composition of the amygdala impedes translational research aimed at developing new treatments and interventions. The aim of this study was to characterize and compare the composition of amygdala cells to help bridge the gap between preclinical models and human psychiatric and neurodevelopmental disorders. METHODS Tissue was dissected from multiple amygdala subnuclei in both humans (N=3, male) and rhesus macaques (N=3, male). Single-nucleus RNA sequencing was performed to characterize the transcriptomes of individual nuclei. RESULTS The results reveal substantial heterogeneity between regions, even when restricted to inhibitory or excitatory neurons. Consistent with previous work, the data highlight the complexities of individual marker genes for uniquely targeting specific cell types. Cross-species analyses suggest that the rhesus monkey model is well-suited to understanding the human amygdala, but also identify limitations. For example, a cell cluster in the ventral lateral nucleus of the amygdala (vLa) is enriched in humans relative to rhesus macaques. Additionally, the data describe specific cell clusters with relative enrichment of disorder-related genes. These analyses point to the human-enriched vLa cell cluster as relevant to autism spectrum disorder, potentially highlighting a vulnerability to neurodevelopmental disorders that has emerged in recent primate evolution. Further, a cluster of cells expressing markers for intercalated cells is enriched for genes reported in human genome-wide association studies of neuroticism, anxiety disorders, and depressive disorders. CONCLUSIONS Together, these findings shed light on the composition of the amygdala and identify specific cell types that can be prioritized in basic science research to better understand human psychopathology and guide the development of potential treatments.
Collapse
Affiliation(s)
- Shawn Kamboj
- Department of Psychology (Kamboj, Fox), California National Primate Research Center (Kamboj, Bauman, Fox), and MIND Institute (Carlson, Ander, Hanson, Bauman, Schumann), University of California, Davis; Department of Psychiatry and Behavioral Sciences (Carlson, Hanson, Schumann), Department of Neurology (Ander), and Department of Physiology and Membrane Biology (Murray, Bauman), School of Medicine, University of California, Davis; Department of Neuroscience and Department of Psychiatry, School of Medicine and Dentistry, University of Rochester, Rochester, NY (Fudge)
| | - Erin L Carlson
- Department of Psychology (Kamboj, Fox), California National Primate Research Center (Kamboj, Bauman, Fox), and MIND Institute (Carlson, Ander, Hanson, Bauman, Schumann), University of California, Davis; Department of Psychiatry and Behavioral Sciences (Carlson, Hanson, Schumann), Department of Neurology (Ander), and Department of Physiology and Membrane Biology (Murray, Bauman), School of Medicine, University of California, Davis; Department of Neuroscience and Department of Psychiatry, School of Medicine and Dentistry, University of Rochester, Rochester, NY (Fudge)
| | - Bradley P Ander
- Department of Psychology (Kamboj, Fox), California National Primate Research Center (Kamboj, Bauman, Fox), and MIND Institute (Carlson, Ander, Hanson, Bauman, Schumann), University of California, Davis; Department of Psychiatry and Behavioral Sciences (Carlson, Hanson, Schumann), Department of Neurology (Ander), and Department of Physiology and Membrane Biology (Murray, Bauman), School of Medicine, University of California, Davis; Department of Neuroscience and Department of Psychiatry, School of Medicine and Dentistry, University of Rochester, Rochester, NY (Fudge)
| | - Kari L Hanson
- Department of Psychology (Kamboj, Fox), California National Primate Research Center (Kamboj, Bauman, Fox), and MIND Institute (Carlson, Ander, Hanson, Bauman, Schumann), University of California, Davis; Department of Psychiatry and Behavioral Sciences (Carlson, Hanson, Schumann), Department of Neurology (Ander), and Department of Physiology and Membrane Biology (Murray, Bauman), School of Medicine, University of California, Davis; Department of Neuroscience and Department of Psychiatry, School of Medicine and Dentistry, University of Rochester, Rochester, NY (Fudge)
| | - Karl D Murray
- Department of Psychology (Kamboj, Fox), California National Primate Research Center (Kamboj, Bauman, Fox), and MIND Institute (Carlson, Ander, Hanson, Bauman, Schumann), University of California, Davis; Department of Psychiatry and Behavioral Sciences (Carlson, Hanson, Schumann), Department of Neurology (Ander), and Department of Physiology and Membrane Biology (Murray, Bauman), School of Medicine, University of California, Davis; Department of Neuroscience and Department of Psychiatry, School of Medicine and Dentistry, University of Rochester, Rochester, NY (Fudge)
| | - Julie L Fudge
- Department of Psychology (Kamboj, Fox), California National Primate Research Center (Kamboj, Bauman, Fox), and MIND Institute (Carlson, Ander, Hanson, Bauman, Schumann), University of California, Davis; Department of Psychiatry and Behavioral Sciences (Carlson, Hanson, Schumann), Department of Neurology (Ander), and Department of Physiology and Membrane Biology (Murray, Bauman), School of Medicine, University of California, Davis; Department of Neuroscience and Department of Psychiatry, School of Medicine and Dentistry, University of Rochester, Rochester, NY (Fudge)
| | - Melissa D Bauman
- Department of Psychology (Kamboj, Fox), California National Primate Research Center (Kamboj, Bauman, Fox), and MIND Institute (Carlson, Ander, Hanson, Bauman, Schumann), University of California, Davis; Department of Psychiatry and Behavioral Sciences (Carlson, Hanson, Schumann), Department of Neurology (Ander), and Department of Physiology and Membrane Biology (Murray, Bauman), School of Medicine, University of California, Davis; Department of Neuroscience and Department of Psychiatry, School of Medicine and Dentistry, University of Rochester, Rochester, NY (Fudge)
| | - Cynthia M Schumann
- Department of Psychology (Kamboj, Fox), California National Primate Research Center (Kamboj, Bauman, Fox), and MIND Institute (Carlson, Ander, Hanson, Bauman, Schumann), University of California, Davis; Department of Psychiatry and Behavioral Sciences (Carlson, Hanson, Schumann), Department of Neurology (Ander), and Department of Physiology and Membrane Biology (Murray, Bauman), School of Medicine, University of California, Davis; Department of Neuroscience and Department of Psychiatry, School of Medicine and Dentistry, University of Rochester, Rochester, NY (Fudge)
| | - Andrew S Fox
- Department of Psychology (Kamboj, Fox), California National Primate Research Center (Kamboj, Bauman, Fox), and MIND Institute (Carlson, Ander, Hanson, Bauman, Schumann), University of California, Davis; Department of Psychiatry and Behavioral Sciences (Carlson, Hanson, Schumann), Department of Neurology (Ander), and Department of Physiology and Membrane Biology (Murray, Bauman), School of Medicine, University of California, Davis; Department of Neuroscience and Department of Psychiatry, School of Medicine and Dentistry, University of Rochester, Rochester, NY (Fudge)
| |
Collapse
|
6
|
Lim H, Zhang Y, Peters C, Straub T, Mayer JL, Klein R. Genetically- and spatially-defined basolateral amygdala neurons control food consumption and social interaction. Nat Commun 2024; 15:6868. [PMID: 39127719 PMCID: PMC11316773 DOI: 10.1038/s41467-024-50889-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
The basolateral amygdala (BLA) contains discrete neuronal circuits that integrate positive or negative emotional information and drive the appropriate innate and learned behaviors. Whether these circuits consist of genetically-identifiable and anatomically segregated neuron types, is poorly understood. Also, our understanding of the response patterns and behavioral spectra of genetically-identifiable BLA neurons is limited. Here, we classified 11 glutamatergic cell clusters in mouse BLA and found that several of them were anatomically segregated in lateral versus basal amygdala, and anterior versus posterior regions of the BLA. Two of these BLA subpopulations innately responded to valence-specific, whereas one responded to mixed - aversive and social - cues. Positive-valence BLA neurons promoted normal feeding, while mixed selectivity neurons promoted fear learning and social interactions. These findings enhance our understanding of cell type diversity and spatial organization of the BLA and the role of distinct BLA populations in representing valence-specific and mixed stimuli.
Collapse
Affiliation(s)
- Hansol Lim
- Department Molecules - Signaling - Development, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Yue Zhang
- Department Synapses - Circuits - Plasticity, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Christian Peters
- Department Molecules - Signaling - Development, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Tobias Straub
- Biomedical Center Core Facility Bioinformatics, LMU, Munich, Germany
| | - Johanna Luise Mayer
- Department Molecules - Signaling - Development, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Rüdiger Klein
- Department Molecules - Signaling - Development, Max Planck Institute for Biological Intelligence, Martinsried, Germany.
| |
Collapse
|
7
|
van Velthoven CTJ, Gao Y, Kunst M, Lee C, McMillen D, Chakka AB, Casper T, Clark M, Chakrabarty R, Daniel S, Dolbeare T, Ferrer R, Gloe J, Goldy J, Guzman J, Halterman C, Ho W, Huang M, James K, Nguy B, Pham T, Ronellenfitch K, Thomas ED, Torkelson A, Pagan CM, Kruse L, Dee N, Ng L, Waters J, Smith KA, Tasic B, Yao Z, Zeng H. The transcriptomic and spatial organization of telencephalic GABAergic neuronal types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599583. [PMID: 38948843 PMCID: PMC11212977 DOI: 10.1101/2024.06.18.599583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The telencephalon of the mammalian brain comprises multiple regions and circuit pathways that play adaptive and integrative roles in a variety of brain functions. There is a wide array of GABAergic neurons in the telencephalon; they play a multitude of circuit functions, and dysfunction of these neurons has been implicated in diverse brain disorders. In this study, we conducted a systematic and in-depth analysis of the transcriptomic and spatial organization of GABAergic neuronal types in all regions of the mouse telencephalon and their developmental origins. This was accomplished by utilizing 611,423 single-cell transcriptomes from the comprehensive and high-resolution transcriptomic and spatial cell type atlas for the adult whole mouse brain we have generated, supplemented with an additional single-cell RNA-sequencing dataset containing 99,438 high-quality single-cell transcriptomes collected from the pre- and postnatal developing mouse brain. We present a hierarchically organized adult telencephalic GABAergic neuronal cell type taxonomy of 7 classes, 52 subclasses, 284 supertypes, and 1,051 clusters, as well as a corresponding developmental taxonomy of 450 clusters across different ages. Detailed charting efforts reveal extraordinary complexity where relationships among cell types reflect both spatial locations and developmental origins. Transcriptomically and developmentally related cell types can often be found in distant and diverse brain regions indicating that long-distance migration and dispersion is a common characteristic of nearly all classes of telencephalic GABAergic neurons. Additionally, we find various spatial dimensions of both discrete and continuous variations among related cell types that are correlated with gene expression gradients. Lastly, we find that cortical, striatal and some pallidal GABAergic neurons undergo extensive postnatal diversification, whereas septal and most pallidal GABAergic neuronal types emerge simultaneously during the embryonic stage with limited postnatal diversification. Overall, the telencephalic GABAergic cell type taxonomy can serve as a foundational reference for molecular, structural and functional studies of cell types and circuits by the entire community.
Collapse
Affiliation(s)
| | - Yuan Gao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mike Huang
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Beagan Nguy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
8
|
Wang M, Luo J, Dai L, Feng M, Cao X, Zhang J, Wan Y, Yang X, Wang Y. Foxp2 deficiency impairs reproduction by modulating the hypothalamic-pituitary-gonadal axis in zebrafish†. Biol Reprod 2024; 110:908-923. [PMID: 38288660 DOI: 10.1093/biolre/ioae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/29/2024] Open
Abstract
FOXP2 was initially characterized as a transcription factor linked to speech and language disorders. Single-cell RNA sequencing reveals that Foxp2 is enriched in the gonadotrope cluster of the pituitary gland and colocalized with the hormones LHB and FSHB in chickens and mice, implying that FOXP2 might be associated with reproduction in vertebrates. Herein, we investigated the roles of foxp2 in reproduction in a Foxp2-deficient zebrafish model. The results indicated that the loss of Foxp2 inhibits courtship behavior in adult male zebrafish. Notably, Foxp2 deficiency disrupts gonad development, leading to retardation of follicle development and a decrease in oocytes in females at the full-growth stage, among other phenotypes. The transcriptome analysis (RNA-seq) also revealed that differentially expressed genes clustered into the estrogen signaling and ovarian steroidogenesis-related signaling pathways. In addition, we found that Foxp2 deficiency could modulate the hypothalamic-pituitary-gonadal axis, especially the regulation of lhb and fshb expression, in zebrafish. In contrast, the injection of human chorionic gonadotropin, a specific LH agonist, partially rescues Foxp2-impaired reproduction in zebrafish, suggesting that Foxp2 plays an important role in the regulation of reproduction via the hypothalamic-pituitary-gonadal axis in zebrafish. Thus, our findings reveal a new role for Foxp2 in the regulation of reproduction in vertebrates.
Collapse
Affiliation(s)
- Maya Wang
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Juanjuan Luo
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Lu Dai
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Meilan Feng
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Xiaoqian Cao
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Jiannan Zhang
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Yiping Wan
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Xiaojun Yang
- Shantou University Medical College, Shantou, China
| | - Yajun Wang
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Alderman PJ, Saxon D, Torrijos-Saiz LI, Sharief M, Page CE, Baroudi JK, Biagiotti SW, Butyrkin VA, Melamed A, Kuo CT, Vicini S, García-Verdugo JM, Herranz-Pérez V, Corbin JG, Sorrells SF. Delayed maturation and migration of excitatory neurons in the juvenile mouse paralaminar amygdala. Neuron 2024; 112:574-592.e10. [PMID: 38086370 PMCID: PMC10922384 DOI: 10.1016/j.neuron.2023.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/05/2023] [Accepted: 11/09/2023] [Indexed: 02/12/2024]
Abstract
The human amygdala paralaminar nucleus (PL) contains many immature excitatory neurons that undergo prolonged maturation from birth to adulthood. We describe a previously unidentified homologous PL region in mice that contains immature excitatory neurons and has previously been considered part of the amygdala intercalated cell clusters or ventral endopiriform cortex. Mouse PL neurons are born embryonically, not from postnatal neurogenesis, despite a subset retaining immature molecular and morphological features in adults. During juvenile-adolescent ages (P21-P35), the majority of PL neurons undergo molecular, structural, and physiological maturation, and a subset of excitatory PL neurons migrate into the adjacent endopiriform cortex. Alongside these changes, PL neurons develop responses to aversive and appetitive olfactory stimuli. The presence of this homologous region in both humans and mice points to the significance of this conserved mechanism of neuronal maturation and migration during adolescence, a key time period for amygdala circuit maturation and related behavioral changes.
Collapse
Affiliation(s)
- Pia J Alderman
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - David Saxon
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20011, USA; Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Lucía I Torrijos-Saiz
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Comparative Neurobiology, University of Valencia, CIBERNED-ISCIII, Valencia 46980, Spain
| | - Malaz Sharief
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Chloe E Page
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Jude K Baroudi
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Sean W Biagiotti
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Vladimir A Butyrkin
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20011, USA; Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD 20742, USA
| | - Anna Melamed
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Chay T Kuo
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Stefano Vicini
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20007, USA; Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Jose M García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Comparative Neurobiology, University of Valencia, CIBERNED-ISCIII, Valencia 46980, Spain; Department of Cell Biology, Functional Biology and Physical Anthropology, University of Valencia, Burjassot 46100, Spain
| | - Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Comparative Neurobiology, University of Valencia, CIBERNED-ISCIII, Valencia 46980, Spain; Department of Cell Biology, Functional Biology and Physical Anthropology, University of Valencia, Burjassot 46100, Spain
| | - Joshua G Corbin
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC 20011, USA
| | - Shawn F Sorrells
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
10
|
Ko BS, Han MH, Kwon MJ, Cha DG, Ji Y, Park ES, Jeon MJ, Kim S, Lee K, Choi YH, Lee J, Torras-Llort M, Yoon KJ, Lee H, Kim JK, Lee SB. Baf-mediated transcriptional regulation of teashirt is essential for the development of neural progenitor cell lineages. Exp Mol Med 2024; 56:422-440. [PMID: 38374207 PMCID: PMC10907700 DOI: 10.1038/s12276-024-01169-3] [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: 03/10/2023] [Revised: 09/20/2023] [Accepted: 12/10/2023] [Indexed: 02/21/2024] Open
Abstract
Accumulating evidence hints heterochromatin anchoring to the inner nuclear membrane as an upstream regulatory process of gene expression. Given that the formation of neural progenitor cell lineages and the subsequent maintenance of postmitotic neuronal cell identity critically rely on transcriptional regulation, it seems possible that the development of neuronal cells is influenced by cell type-specific and/or context-dependent programmed regulation of heterochromatin anchoring. Here, we explored this possibility by genetically disrupting the evolutionarily conserved barrier-to-autointegration factor (Baf) in the Drosophila nervous system. Through single-cell RNA sequencing, we demonstrated that Baf knockdown induces prominent transcriptomic changes, particularly in type I neuroblasts. Among the differentially expressed genes, our genetic analyses identified teashirt (tsh), a transcription factor that interacts with beta-catenin, to be closely associated with Baf knockdown-induced phenotypes that were suppressed by the overexpression of tsh or beta-catenin. We also found that Baf and tsh colocalized in a region adjacent to heterochromatin in type I NBs. Notably, the subnuclear localization pattern remained unchanged when one of these two proteins was knocked down, indicating that both proteins contribute to the anchoring of heterochromatin to the inner nuclear membrane. Overall, this study reveals that the Baf-mediated transcriptional regulation of teashirt is a novel molecular mechanism that regulates the development of neural progenitor cell lineages.
Collapse
Affiliation(s)
- Byung Su Ko
- Department of Brain Sciences, DGIST, Daegu, 42988, Republic of Korea
| | - Myeong Hoon Han
- Department of Brain Sciences, DGIST, Daegu, 42988, Republic of Korea
| | - Min Jee Kwon
- Department of Brain Sciences, DGIST, Daegu, 42988, Republic of Korea
| | - Dong Gon Cha
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Yuri Ji
- Department of Brain Sciences, DGIST, Daegu, 42988, Republic of Korea
| | - Eun Seo Park
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Min Jae Jeon
- Department of Brain Sciences, DGIST, Daegu, 42988, Republic of Korea
| | - Somi Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Kyeongho Lee
- Department of Brain Sciences, DGIST, Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, DGIST, Daegu, 42988, Republic of Korea
| | - Yoon Ha Choi
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Jusung Lee
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | | | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hyosang Lee
- Department of Brain Sciences, DGIST, Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, DGIST, Daegu, 42988, Republic of Korea
| | - Jong Kyoung Kim
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea.
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea.
| | - Sung Bae Lee
- Department of Brain Sciences, DGIST, Daegu, 42988, Republic of Korea.
| |
Collapse
|
11
|
Iyyanar PPR, Qin C, Adhikari N, Liu H, Hu YC, Jiang R, Lan Y. Developmental origin of the mammalian premaxilla. Dev Biol 2023; 503:1-9. [PMID: 37524195 PMCID: PMC10528123 DOI: 10.1016/j.ydbio.2023.07.005] [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: 04/18/2023] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
The evolution of jaws has played a major role in the success of vertebrate expansion into a wide variety of ecological niches. A fundamental, yet unresolved, question in craniofacial biology is about the origin of the premaxilla, the most distal bone present in the upper jaw of all amniotes. Recent reports have suggested that the mammalian premaxilla is derived from embryonic maxillary prominences rather than the frontonasal ectomesenchyme as previously shown in studies of chicken embryos. However, whether mammalian embryonic frontonasal ectomesenchyme contributes to the premaxillary bone has not been investigated and a tool to trace the contributions of the frontonasal ectomesenchyme to facial structures in mammals is lacking. The expression of the Alx3 gene is activated highly specifically in the frontonasal ectomesenchyme, but not in the maxillary mesenchyme, from the beginning of facial morphogenesis in mice. Here, we report the generation and characterization of a novel Alx3CreERT2 knock-in mouse line that express tamoxifen-inducible Cre DNA recombinase from the Alx3 locus. Tamoxifen treatment of Alx3CreERT2/+;Rosa26mTmG/+ embryos at E7.5, E8.5, E9.5, and E10.5, each induced specific labeling of the embryonic medial nasal and lateral nasal mesenchyme but not the maxillary mesenchyme. Lineage tracing of Alx3CreERT2-labeled frontonasal mesenchyme from E9.5 to E16.5 clearly showed that the frontonasal mesenchyme cells give rise to the osteoblasts generating the premaxillary bone. Furthermore, we characterize a Dlx1-Cre BAC transgenic mouse line that expresses Cre activity in the embryonic maxillary but not the frontonasal mesenchyme and show that the Dlx1-Cre labeled embryonic maxillary mesenchyme cells contribute to the maxillary bone as well as the soft tissues lateral to both the premaxillary and maxillary bones but not to the premaxillary bone. These results clearly demonstrate the developmental origin of the premaxillary bone from embryonic frontonasal ectomesenchyme cells in mice and confirm the evolutionary homology of the premaxilla across amniotes.
Collapse
Affiliation(s)
- Paul P R Iyyanar
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Chuanqi Qin
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Ministry of Education Key Laboratory of Oral Biomedicine, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Nirpesh Adhikari
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Han Liu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Yueh-Chiang Hu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Rulang Jiang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| | - Yu Lan
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| |
Collapse
|
12
|
Stern DB, Wilke A, Root CM. Anatomical Connectivity of the Intercalated Cells of the Amygdala. eNeuro 2023; 10:ENEURO.0238-23.2023. [PMID: 37775310 PMCID: PMC10576262 DOI: 10.1523/eneuro.0238-23.2023] [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: 07/06/2023] [Revised: 09/05/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023] Open
Abstract
The intercalated cells of the amygdala (ITCs) are a fundamental processing structure in the amygdala that remain relatively understudied. They are phylogenetically conserved from insectivores through primates, inhibitory, and project to several of the main processing and output stations of the amygdala and basal forebrain. Through these connections, the ITCs are best known for their role in conditioned fear, where they are required for fear extinction learning and recall. Prior work on ITC connectivity is limited, and thus holistic characterization of their afferent and efferent connectivity in a genetically defined manner is incomplete. The ITCs express the FoxP2 transcription factor, affording genetic access to these neurons for viral input-output mapping. To fully characterize the anatomic connectivity of the ITCs, we used cre-dependent viral strategies in FoxP2-cre mice to reveal the projections of the main (mITC), caudal (cITC), and lateral (lITC) clusters along with their presynaptic sources of innervation. Broadly, the results confirm many known pathways, reveal previously unknown ones, and demonstrate important novel insights about each nucleus's unique connectivity profile and relative distributions. We show that the ITCs receive information from a wide range of cortical, subcortical, basal, amygdalar, hippocampal, and thalamic structures, and project broadly to areas of the basal forebrain, hypothalamus, and entire extent of the amygdala. The results provide a comprehensive map of their connectivity and suggest that the ITCs could potentially influence a broad range of behaviors by integrating information from a wide array of sources throughout the brain.
Collapse
Affiliation(s)
- Daniel B Stern
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093
- Neuroscience Graduate Program, University of California, San Diego, La Jolla, CA 92093
| | - Anna Wilke
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093
| | - Cory M Root
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
13
|
Yang S, Zhu G. Phytotherapy of abnormality of fear memory: A narrative review of mechanisms. Fitoterapia 2023; 169:105618. [PMID: 37482307 DOI: 10.1016/j.fitote.2023.105618] [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: 05/23/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
It is generally believed that in post-traumatic stress disorder (PTSD), the high expression of fear memory is mainly determined by amygdala hyperactivity and hippocampus hypoactivity. In this review, we firstly updated the mechanisms of fear memory, and then searched the experimental evidence of phytotherapy for fear memory in the past five years. Based on the summary of those experimental studies, we further discussed the future research strategies of plant medicines, including the study of the mechanism of specific brain regions, the optimal time for the prevention and treatment of fear memory-related diseases such as PTSD, and the development of new drugs with active components of plant medicines. Accordingly, plant medicines play a clear role in improving fear memory abnormalities and have the drug development potential in the treatment of fear-related disorders.
Collapse
Affiliation(s)
- Shaojie Yang
- The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230061, China; Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui 230012, China.
| |
Collapse
|
14
|
Peters C, He S, Fermani F, Lim H, Ding W, Mayer C, Klein R. Transcriptomics reveals amygdala neuron regulation by fasting and ghrelin thereby promoting feeding. SCIENCE ADVANCES 2023; 9:eadf6521. [PMID: 37224253 DOI: 10.1126/sciadv.adf6521] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 04/19/2023] [Indexed: 05/26/2023]
Abstract
The central amygdala (CeA) consists of numerous genetically defined inhibitory neurons that control defensive and appetitive behaviors including feeding. Transcriptomic signatures of cell types and their links to function remain poorly understood. Using single-nucleus RNA sequencing, we describe nine CeA cell clusters, of which four are mostly associated with appetitive and two with aversive behaviors. To analyze the activation mechanism of appetitive CeA neurons, we characterized serotonin receptor 2a (Htr2a)-expressing neurons (CeAHtr2a) that comprise three appetitive clusters and were previously shown to promote feeding. In vivo calcium imaging revealed that CeAHtr2a neurons are activated by fasting, the hormone ghrelin, and the presence of food. Moreover, these neurons are required for the orexigenic effects of ghrelin. Appetitive CeA neurons responsive to fasting and ghrelin project to the parabrachial nucleus (PBN) causing inhibition of target PBN neurons. These results illustrate how the transcriptomic diversification of CeA neurons relates to fasting and hormone-regulated feeding behavior.
Collapse
Affiliation(s)
- Christian Peters
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Songwei He
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Federica Fermani
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Hansol Lim
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Wenyu Ding
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Christian Mayer
- Laboratory of Neurogenomics, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Rüdiger Klein
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| |
Collapse
|
15
|
Molecular and cellular evolution of the amygdala across species analyzed by single-nucleus transcriptome profiling. Cell Discov 2023; 9:19. [PMID: 36788214 PMCID: PMC9929086 DOI: 10.1038/s41421-022-00506-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/24/2022] [Indexed: 02/16/2023] Open
Abstract
The amygdala, or an amygdala-like structure, is found in the brains of all vertebrates and plays a critical role in survival and reproduction. However, the cellular architecture of the amygdala and how it has evolved remain elusive. Here, we generated single-nucleus RNA-sequencing data for more than 200,000 cells in the amygdala of humans, macaques, mice, and chickens. Abundant neuronal cell types from different amygdala subnuclei were identified in all datasets. Cross-species analysis revealed that inhibitory neurons and inhibitory neuron-enriched subnuclei of the amygdala were well-conserved in cellular composition and marker gene expression, whereas excitatory neuron-enriched subnuclei were relatively divergent. Furthermore, LAMP5+ interneurons were much more abundant in primates, while DRD2+ inhibitory neurons and LAMP5+SATB2+ excitatory neurons were dominant in the human central amygdalar nucleus (CEA) and basolateral amygdalar complex (BLA), respectively. We also identified CEA-like neurons and their species-specific distribution patterns in chickens. This study highlights the extreme cell-type diversity in the amygdala and reveals the conservation and divergence of cell types and gene expression patterns across species that may contribute to species-specific adaptations.
Collapse
|
16
|
Dorofeikova M, Borkar CD, Weissmuller K, Smith-Osborne L, Basavanhalli S, Bean E, Smith A, Duong A, Resendez A, Fadok JP. Effects of footshock stress on social behavior and neuronal activation in the medial prefrontal cortex and amygdala of male and female mice. PLoS One 2023; 18:e0281388. [PMID: 36757923 PMCID: PMC9910713 DOI: 10.1371/journal.pone.0281388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 01/21/2023] [Indexed: 02/10/2023] Open
Abstract
Social behavior is complex and fundamental, and its deficits are common pathological features for several psychiatric disorders including anxiety, depression, and posttraumatic stress disorder. Acute stress may have a negative impact on social behavior, and these effects can vary based on sex. The aim of this study was to explore the effect of acute footshock stress, using analogous parameters to those commonly used in fear conditioning assays, on the sociability of male and female C57BL/6J mice in a standard social approach test. Animals were divided into two main groups of footshock stress (22 male, 24 female) and context exposed control (23 male and 22 female). Each group had mice that were treated intraperitoneally with either the benzodiazepine-alprazolam (control: 10 male, 10 female; stress: 11 male, 11 female), or vehicle (control: 13 male, 12 female; stress: 11 male, 13 female). In all groups, neuronal activation during social approach was assessed using immunohistochemistry against the immediate early gene product cFos. Although footshock stress did not significantly alter sociability or latency to approach a social stimulus, it did increase defensive tail-rattling behavior specifically in males (p = 0.0022). This stress-induced increase in tail-rattling was alleviated by alprazolam (p = 0.03), yet alprazolam had no effect on female tail-rattling behavior in the stress group. Alprazolam lowered cFos expression in the medial prefrontal cortex (p = 0.001 infralimbic area, p = 0.02 prelimbic area), and social approach induced sex-dependent differences in cFos activation in the ventromedial intercalated cell clusters (p = 0.04). Social approach following stress-induced cFos expression was positively correlated with latency to approach and negatively correlated with sociability in the prelimbic area and multiple amygdala subregions (all p < 0.05). Collectively, our results suggest that acute footshock stress induces sex-dependent alterations in defensiveness and differential patterns of cFos activation during social approach.
Collapse
Affiliation(s)
- Mariia Dorofeikova
- Department of Psychology, Tulane University, New Orleans, LA, United States of America
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | - Chandrashekhar D. Borkar
- Department of Psychology, Tulane University, New Orleans, LA, United States of America
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | | | - Lydia Smith-Osborne
- Department of Psychology, Tulane University, New Orleans, LA, United States of America
- Tulane National Primate Research Center, Covington, LA, United States of America
| | - Samhita Basavanhalli
- Neuroscience Program, Tulane University, New Orleans, LA, United States of America
| | - Erin Bean
- Neuroscience Program, Tulane University, New Orleans, LA, United States of America
| | - Avery Smith
- Neuroscience Program, Tulane University, New Orleans, LA, United States of America
| | - Anh Duong
- Department of Psychology, Tulane University, New Orleans, LA, United States of America
- Neuroscience Program, Tulane University, New Orleans, LA, United States of America
| | - Alexis Resendez
- Department of Psychology, Tulane University, New Orleans, LA, United States of America
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | - Jonathan P. Fadok
- Department of Psychology, Tulane University, New Orleans, LA, United States of America
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
- * E-mail:
| |
Collapse
|
17
|
Deryckere A, Woych J, Jaeger ECB, Tosches MA. Molecular Diversity of Neuron Types in the Salamander Amygdala and Implications for Amygdalar Evolution. BRAIN, BEHAVIOR AND EVOLUTION 2022; 98:61-75. [PMID: 36574764 PMCID: PMC10096051 DOI: 10.1159/000527899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/21/2022] [Indexed: 12/28/2022]
Abstract
The amygdala is a complex brain structure in the vertebrate telencephalon, essential for regulating social behaviors, emotions, and (social) cognition. In contrast to the vast majority of neuron types described in the many nuclei of the mammalian amygdala, little is known about the neuronal diversity in non-mammals, making reconstruction of its evolution particularly difficult. Here, we characterize glutamatergic neuron types in the amygdala of the urodele amphibian Pleurodeles waltl. Our single-cell RNA sequencing data indicate the existence of at least ten distinct types and subtypes of glutamatergic neurons in the salamander amygdala. These neuron types are molecularly distinct from neurons in the ventral pallium (VP), suggesting that the pallial amygdala and the VP are two separate areas in the telencephalon. In situ hybridization for marker genes indicates that amygdalar glutamatergic neuron types are located in three major subdivisions: the lateral amygdala, the medial amygdala, and a newly defined area demarcated by high expression of the transcription factor Sim1. The gene expression profiles of these neuron types suggest similarities with specific neurons in the sauropsid and mammalian amygdala. In particular, we identify Sim1+ and Sim1+ Otp+ expressing neuron types, potentially homologous to the mammalian nucleus of the lateral olfactory tract (NLOT) and to hypothalamic-derived neurons of the medial amygdala, respectively. Taken together, our results reveal a surprising diversity of glutamatergic neuron types in the amygdala of salamanders, despite the anatomical simplicity of their brain. These results offer new insights on the cellular and anatomical complexity of the amygdala in tetrapod ancestors.
Collapse
Affiliation(s)
- Astrid Deryckere
- Department of Biological Sciences, Columbia University; New York, NY 10027, USA
| | - Jamie Woych
- Department of Biological Sciences, Columbia University; New York, NY 10027, USA
| | - Eliza C. B. Jaeger
- Department of Biological Sciences, Columbia University; New York, NY 10027, USA
| | | |
Collapse
|
18
|
Asede D, Doddapaneni D, Bolton MM. Amygdala Intercalated Cells: Gate Keepers and Conveyors of Internal State to the Circuits of Emotion. J Neurosci 2022; 42:9098-9109. [PMID: 36639901 PMCID: PMC9761677 DOI: 10.1523/jneurosci.1176-22.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/19/2022] [Accepted: 10/16/2022] [Indexed: 01/09/2023] Open
Abstract
Generating adaptive behavioral responses to emotionally salient stimuli requires evaluation of complex associations between multiple sensations, the surrounding context, and current internal state. Neural circuits within the amygdala parse this emotional information, undergo synaptic plasticity to reflect learned associations, and evoke appropriate responses through their projections to the brain regions orchestrating these behaviors. Information flow within the amygdala is regulated by the intercalated cells (ITCs), which are densely packed clusters of GABAergic neurons that encircle the basolateral amygdala (BLA) and provide contextually relevant feedforward inhibition of amygdala nuclei, including the central and BLA. Emerging studies have begun to delineate the unique contribution of each ITC cluster and establish ITCs as key loci of plasticity in emotional learning. In this review, we summarize the known connectivity and function of individual ITC clusters and explore how different neuromodulators conveying internal state act via ITC gates to shape emotionally motivated behavior. We propose that the behavioral state-dependent function of ITCs, their unique genetic profile, and rich expression of neuromodulator receptors make them potential therapeutic targets for disorders, such as anxiety, schizophrenia spectrum, and addiction.
Collapse
Affiliation(s)
- Douglas Asede
- Disorders of Neural Circuit Function, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Divyesh Doddapaneni
- Disorders of Neural Circuit Function, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - M McLean Bolton
- Disorders of Neural Circuit Function, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| |
Collapse
|
19
|
Dilly GA, Kittleman CW, Kerr TM, Messing RO, Mayfield RD. Cell-type specific changes in PKC-delta neurons of the central amygdala during alcohol withdrawal. Transl Psychiatry 2022; 12:289. [PMID: 35859068 PMCID: PMC9300707 DOI: 10.1038/s41398-022-02063-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 02/08/2023] Open
Abstract
The central amygdala (CeA) contains a diverse population of cells, including multiple subtypes of GABAergic neurons, along with glia and epithelial cells. Specific CeA cell types have been shown to affect alcohol consumption in animal models of dependence and may be involved in negative affect during alcohol withdrawal. We used single-nuclei RNA sequencing to determine cell-type specificity of differential gene expression in the CeA induced by alcohol withdrawal. Cells within the CeA were classified using unbiased clustering analyses and identified based on the expression of known marker genes. Differential gene expression analysis was performed on each identified CeA cell-type. It revealed differential gene expression in astrocytes and GABAergic neurons associated with alcohol withdrawal. GABAergic neurons were further subclassified into 13 clusters of cells. Analyzing transcriptomic responses in these subclusters revealed that alcohol exposure induced multiple differentially expressed genes in one subtype of CeA GABAergic neurons, the protein kinase C delta (PKCδ) expressing neurons. These results suggest that PKCδ neurons in the CeA may be uniquely sensitive to the effects of alcohol exposure and identify a novel population of cells in CeA associated with alcohol withdrawal.
Collapse
Affiliation(s)
- Geoffrey A. Dilly
- grid.89336.370000 0004 1936 9924Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Department of Neurology, The University of Texas at Austin, Austin, TX 78712 USA
| | - Cory W. Kittleman
- grid.89336.370000 0004 1936 9924Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712 USA
| | - Tony M. Kerr
- grid.89336.370000 0004 1936 9924Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Department of Neurology, The University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924College of Pharmacy, The University of Texas at Austin, Austin, TX 78712 USA
| | - Robert O. Messing
- grid.89336.370000 0004 1936 9924Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Department of Neurology, The University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924College of Pharmacy, The University of Texas at Austin, Austin, TX 78712 USA
| | - R. Dayne Mayfield
- grid.89336.370000 0004 1936 9924Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712 USA ,grid.89336.370000 0004 1936 9924Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712 USA
| |
Collapse
|
20
|
Biggs LM, Meredith M. Functional connectivity of intercalated nucleus with medial amygdala: A circuit relevant for chemosignal processing. IBRO Neurosci Rep 2022; 12:170-181. [PMID: 35199098 PMCID: PMC8850325 DOI: 10.1016/j.ibneur.2022.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/08/2022] [Accepted: 01/19/2022] [Indexed: 11/21/2022] Open
Abstract
Medial amygdala processes social/reproductive chemosensory input, and its projections to preoptic and hypothalamic areas evoke appropriate behavioral and physiological responses. We and others have shown that different chemosensory signals elicit differential responses in medial amygdala subregions and in adjacent main intercalated nucleus (mICN). The largely GABAergic mICN receives no direct chemosensory input but, as we show, mICN has functional circuit connections with medial amygdala that could be responsible both for mICN chemosensitivity and for a feedforward inhibitory effect on posterior medial amygdala; which, in turn would affect chemosignal response. mICN is subject to inhibition by dopamine and is probably regulated by neuropeptides and input from frontal cortex. Thus, mICN is in position to modify chemosensory processing in medial amygdala and behavioral responses to social signals, according to internal brain state. Patch-clamp recordings from neurons in each relevant nucleus in horizontal brain-slices, with electrical stimulation in adjacent nuclei, reveal multiple functional connections between medial amygdala subregions and mICN. We highlight a triangular circuit which may underlie mICN chemosensitivity and its potential for modifying chemosensory information transmitted to basal forebrain. Anterior medial amygdala, which receives most of the chemosensory input, connects to posterior medial amygdala directly and both areas send information on to basal forebrain. Anterior medial amygdala can also modulate posterior medial amygdala indirectly via the mICN side-loop, which also provides a pathway for modulation by cortical input or, when inhibited by dopamine, could allow a more automatic response - as proposed for other amygdala circuits with similar ICN side loops.
Collapse
Affiliation(s)
| | - Michael Meredith
- Program in Neuroscience and Dept. Biological Science, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
21
|
Schmitz MT, Sandoval K, Chen CP, Mostajo-Radji MA, Seeley WW, Nowakowski TJ, Ye CJ, Paredes MF, Pollen AA. The development and evolution of inhibitory neurons in primate cerebrum. Nature 2022; 603:871-877. [PMID: 35322231 PMCID: PMC8967711 DOI: 10.1038/s41586-022-04510-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 02/01/2022] [Indexed: 12/14/2022]
Abstract
Neuroanatomists have long speculated that expanded primate brains contain an increased morphological diversity of inhibitory neurons (INs)1, and recent studies have identified primate-specific neuronal populations at the molecular level2. However, we know little about the developmental mechanisms that specify evolutionarily novel cell types in the brain. Here, we reconstruct gene expression trajectories specifying INs generated throughout the neurogenic period in macaques and mice by analysing the transcriptomes of 250,181 cells. We find that the initial classes of INs generated prenatally are largely conserved among mammals. Nonetheless, we identify two contrasting developmental mechanisms for specifying evolutionarily novel cell types during prenatal development. First, we show that recently identified primate-specific TAC3 striatal INs are specified by a unique transcriptional programme in progenitors followed by induction of a distinct suite of neuropeptides and neurotransmitter receptors in new-born neurons. Second, we find that multiple classes of transcriptionally conserved olfactory bulb (OB)-bound precursors are redirected to expanded primate white matter and striatum. These classes include a novel peristriatal class of striatum laureatum neurons that resemble dopaminergic periglomerular cells of the OB. We propose an evolutionary model in which conserved initial classes of neurons supplying the smaller primate OB are reused in the enlarged striatum and cortex. Together, our results provide a unified developmental taxonomy of initial classes of mammalian INs and reveal multiple developmental mechanisms for neural cell type evolution.
Collapse
Affiliation(s)
- Matthew T Schmitz
- Eli and Edythe Broad Center for 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
| | - Kadellyn Sandoval
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher P Chen
- Eli and Edythe Broad Center for 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
| | - Mohammed A Mostajo-Radji
- Eli and Edythe Broad Center for 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
| | - William W Seeley
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Chun Jimmie Ye
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Mercedes F Paredes
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Alex A Pollen
- Eli and Edythe Broad Center for 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.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
22
|
Zoubovsky SP, Williams MT, Hoseus S, Tumukuntala S, Riesenberg A, Schulkin J, Vorhees CV, Campbell K, Lim HW, Muglia LJ. Neurobehavioral abnormalities following prenatal psychosocial stress are differentially modulated by maternal environment. Transl Psychiatry 2022; 12:22. [PMID: 35039487 PMCID: PMC8764031 DOI: 10.1038/s41398-022-01785-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 12/20/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
Prenatal stress (PS) is associated with increased vulnerability to affective disorders. Transplacental glucocorticoid passage and stress-induced maternal environment alterations are recognized as potential routes of transmission that can fundamentally alter neurodevelopment. However, molecular mechanisms underlying aberrant emotional outcomes or the individual contributions intrauterine stress versus maternal environment play in shaping these mechanisms remain unknown. Here, we report anxiogenic behaviors, anhedonia, and female hypothalamic-pituitary-adrenal axis hyperactivity as a consequence of psychosocial PS in mice. Evidence of fetal amygdala programming precedes these abnormalities. In adult offspring, we observe amygdalar transcriptional changes demonstrating sex-specific dysfunction in synaptic transmission and neurotransmitter systems. We find these abnormalities are primarily driven by in-utero stress exposure. Importantly, maternal care changes postnatally reverse anxiety-related behaviors and partially rescue gene alterations associated with neurotransmission. Our data demonstrate the influence maternal environment exerts in shaping offspring emotional development despite deleterious effects of intrauterine stress.
Collapse
Affiliation(s)
- Sandra P. Zoubovsky
- grid.24827.3b0000 0001 2179 9593Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Michael T. Williams
- grid.24827.3b0000 0001 2179 9593Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Sarah Hoseus
- grid.239573.90000 0000 9025 8099Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Shivani Tumukuntala
- grid.239573.90000 0000 9025 8099Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Amy Riesenberg
- grid.239573.90000 0000 9025 8099Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Jay Schulkin
- grid.213910.80000 0001 1955 1644Department of Neuroscience, Georgetown University, Washington, DC USA ,grid.34477.330000000122986657Department of Obstetrics and Gynecology, University of Washington, Seattle, WA USA
| | - Charles V. Vorhees
- grid.24827.3b0000 0001 2179 9593Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Kenneth Campbell
- grid.24827.3b0000 0001 2179 9593Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Division of Neurosurgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Hee-Woong Lim
- grid.24827.3b0000 0001 2179 9593Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Louis J. Muglia
- grid.24827.3b0000 0001 2179 9593Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.427464.70000 0000 8727 8697Office of the President, Burroughs Wellcome Fund, Research Triangle Park, NC USA
| |
Collapse
|
23
|
Aerts T, Seuntjens E. Novel Perspectives on the Development of the Amygdala in Rodents. Front Neuroanat 2021; 15:786679. [PMID: 34955766 PMCID: PMC8696165 DOI: 10.3389/fnana.2021.786679] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022] Open
Abstract
The amygdala is a hyperspecialized brain region composed of strongly inter- and intraconnected nuclei involved in emotional learning and behavior. The cellular heterogeneity of the amygdalar nuclei has complicated straightforward conclusions on their developmental origin, and even resulted in contradictory data. Recently, the concentric ring theory of the pallium and the radial histogenetic model of the pallial amygdala have cleared up several uncertainties that plagued previous models of amygdalar development. Here, we provide an extensive overview on the developmental origin of the nuclei of the amygdaloid complex. Starting from older gene expression data, transplantation and lineage tracing studies, we systematically summarize and reinterpret previous findings in light of the novel perspectives on amygdalar development. In addition, migratory routes that these cells take on their way to the amygdala are explored, and known transcription factors and guidance cues that seemingly drive these cells toward the amygdala are emphasized. We propose some future directions for research on amygdalar development and highlight that a better understanding of its development could prove critical for the treatment of several neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tania Aerts
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Eve Seuntjens
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Xu S, Jiang M, Liu X, Sun Y, Yang L, Yang Q, Bai Z. Neural Circuits for Social Interactions: From Microcircuits to Input-Output Circuits. Front Neural Circuits 2021; 15:768294. [PMID: 34776877 PMCID: PMC8585935 DOI: 10.3389/fncir.2021.768294] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/11/2021] [Indexed: 11/20/2022] Open
Abstract
Social behaviors entail responses to social information and requires the perception and integration of social cues through a complex cognition process that involves attention, memory, motivation, and emotion. Neurobiological and molecular mechanisms underlying social behavior are highly conserved across species, and inter- and intra-specific variability observed in social behavior can be explained to large extent by differential activity of a conserved neural network. However, neural microcircuits and precise networks involved in social behavior remain mysterious. In this review, we summarize the microcircuits and input-output circuits on the molecular, cellular, and network levels of different social interactions, such as social exploration, social hierarchy, social memory, and social preference. This review provides a broad view of how multiple microcircuits and input-output circuits converge on the medial prefrontal cortex, hippocampus, and amygdala to regulate complex social behaviors, as well as a potential novel view for better control over pathological development.
Collapse
Affiliation(s)
- Sen Xu
- Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, College of Life Sciences and Research Center for Resource Peptide Drugs, Yanan University, Yanan, China
| | - Ming Jiang
- Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, College of Life Sciences and Research Center for Resource Peptide Drugs, Yanan University, Yanan, China
| | - Xia Liu
- Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, College of Life Sciences and Research Center for Resource Peptide Drugs, Yanan University, Yanan, China
| | - Yahan Sun
- Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, College of Life Sciences and Research Center for Resource Peptide Drugs, Yanan University, Yanan, China
| | - Liang Yang
- Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, College of Life Sciences and Research Center for Resource Peptide Drugs, Yanan University, Yanan, China
| | - Qinghu Yang
- Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, College of Life Sciences and Research Center for Resource Peptide Drugs, Yanan University, Yanan, China
| | - Zhantao Bai
- Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, College of Life Sciences and Research Center for Resource Peptide Drugs, Yanan University, Yanan, China
| |
Collapse
|
25
|
Herrero MJ, Wang L, Hernandez-Pineda D, Banerjee P, Matos HY, Goodrich M, Panigrahi A, Smith NA, Corbin JG. Sex-Specific Social Behavior and Amygdala Proteomic Deficits in Foxp2 +/- Mutant Mice. Front Behav Neurosci 2021; 15:706079. [PMID: 34421555 PMCID: PMC8374433 DOI: 10.3389/fnbeh.2021.706079] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/29/2021] [Indexed: 12/20/2022] Open
Abstract
In humans, mutations in the transcription factor encoding gene, FOXP2, are associated with language and Autism Spectrum Disorders (ASD), the latter characterized by deficits in social interactions. However, little is known regarding the function of Foxp2 in male or female social behavior. Our previous studies in mice revealed high expression of Foxp2 within the medial subnucleus of the amygdala (MeA), a limbic brain region highly implicated in innate social behaviors such as mating, aggression, and parental care. Here, using a comprehensive panel of behavioral tests in male and female Foxp2 +/- heterozygous mice, we investigated the role Foxp2 plays in MeA-linked innate social behaviors. We reveal significant deficits in olfactory processing, social interaction, mating, aggressive, and parental behaviors. Interestingly, some of these deficits are displayed in a sex-specific manner. To examine the consequences of Foxp2 loss of function specifically in the MeA, we conducted a proteomic analysis of microdissected MeA tissue. This analyses revealed putative sex differences expression of a host of proteins implicated in neuronal communication, connectivity, and dopamine signaling. Consistent with this, we discovered that MeA Foxp2-lineage cells were responsive to dopamine with differences between males and females. Thus, our findings reveal a central and sex-specific role for Foxp2 in social behavior and MeA function.
Collapse
Affiliation(s)
- Maria Jesus Herrero
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - Li Wang
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - David Hernandez-Pineda
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - Payal Banerjee
- Center for Genomic Medicine, Children’s National Hospital, Washington, DC, United States
| | - Heidi Y. Matos
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - Meredith Goodrich
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - Aswini Panigrahi
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC, United States
| | - Nathan Anthony Smith
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - Joshua G. Corbin
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| |
Collapse
|
26
|
Aksoy-Aksel A, Gall A, Seewald A, Ferraguti F, Ehrlich I. Midbrain dopaminergic inputs gate amygdala intercalated cell clusters by distinct and cooperative mechanisms in male mice. eLife 2021; 10:e63708. [PMID: 34028352 PMCID: PMC8143799 DOI: 10.7554/elife.63708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 05/06/2021] [Indexed: 01/04/2023] Open
Abstract
Dopaminergic signaling plays an important role in associative learning, including fear and extinction learning. Dopaminergic midbrain neurons encode prediction error-like signals when threats differ from expectations. Within the amygdala, GABAergic intercalated cell (ITC) clusters receive one of the densest dopaminergic projections, but their physiological consequences are incompletely understood. ITCs are important for fear extinction, a function thought to be supported by activation of ventromedial ITCs that inhibit central amygdala fear output. In mice, we reveal two distinct novel mechanisms by which mesencephalic dopaminergic afferents control ITCs. Firstly, they co-release GABA to mediate rapid, direct inhibition. Secondly, dopamine suppresses inhibitory interactions between distinct ITC clusters via presynaptic D1 receptors. Early extinction training augments both GABA co-release onto dorsomedial ITCs and dopamine-mediated suppression of dorso- to ventromedial inhibition between ITC clusters. These findings provide novel insights into dopaminergic mechanisms shaping the activity balance between distinct ITC clusters that could support their opposing roles in fear behavior.
Collapse
Affiliation(s)
- Ayla Aksoy-Aksel
- Hertie Institute for Clinical Brain ResearchTübingenGermany
- Centre for Integrative NeuroscienceTübingenGermany
- Department of Neurobiology, Institute of Biomaterials and Biomolecular Systems, University of StuttgartStuttgartGermany
| | - Andrea Gall
- Hertie Institute for Clinical Brain ResearchTübingenGermany
- Centre for Integrative NeuroscienceTübingenGermany
- Department of Neurobiology, Institute of Biomaterials and Biomolecular Systems, University of StuttgartStuttgartGermany
| | - Anna Seewald
- Department of Pharmacology, Medical University of InnsbruckInnsbruckAustria
| | | | - Ingrid Ehrlich
- Hertie Institute for Clinical Brain ResearchTübingenGermany
- Centre for Integrative NeuroscienceTübingenGermany
- Department of Neurobiology, Institute of Biomaterials and Biomolecular Systems, University of StuttgartStuttgartGermany
| |
Collapse
|
27
|
Apical intercalated cell cluster: A distinct sensory regulator in the amygdala. Cell Rep 2021; 35:109151. [PMID: 34010641 DOI: 10.1016/j.celrep.2021.109151] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 02/20/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
GABAergic neurons regulate different aspects of information processing in the amygdala. Among these are clusters of intercalated cells (ITCs), which have been implicated in fear-related behaviors. Although a few of the ITC clusters have been studied, the functional role of apical ITCs (apITCs) is unknown. Here, we combine monosynaptic rabies tracing with optogenetics and demonstrate that apITCs receive synaptic input from medial geniculate nucleus (MGm), posterior intralaminar nucleus (PIN), and medial dorsal nucleus of the thalamus and from a diverse range of cortical areas including temporal association, entorhinal, insular, piriform, and somatosensory cortex. Upon fear learning, PIN/MGm inputs are strengthened, indicative of their involvement in fear behaviors. 3-D reconstruction of apITCs reveals local arborization and innervation of the dorsal striatum and lateral amygdala. We further show that apITCs provide sensory feedforward inhibition to LA principal cells, a putative mechanism for controlling plasticity during fear learning.
Collapse
|
28
|
Talley MJ, Nardini D, Qin S, Prada CE, Ehrman LA, Waclaw RR. A role for sustained MAPK activity in the mouse ventral telencephalon. Dev Biol 2021; 476:137-147. [PMID: 33775695 DOI: 10.1016/j.ydbio.2021.03.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 03/14/2021] [Accepted: 03/21/2021] [Indexed: 11/28/2022]
Abstract
The MAPK pathway is a major growth signal that has been implicated during the development of progenitors, neurons, and glia in the embryonic brain. Here, we show that the MAPK pathway plays an important role in the generation of distinct cell types from progenitors in the ventral telencephalon. Our data reveal that phospho-p44/42 (called p-ERK1/2) and the ETS transcription factor Etv5, both downstream effectors in the MAPK pathway, show a regional bias in expression during ventral telencephalic development, with enriched expression in the dorsal region of the LGE and ventral region of the MGE at E13.5 and E15.5. Interestingly, expression of both factors becomes more uniform in ventricular zone (VZ) progenitors by E18.5. To gain insight into the role of MAPK activity during progenitor cell development, we used a cre inducible constitutively active MEK1 allele (RosaMEK1DD/+) in combination with a ventral telencephalon enriched cre (Gsx2e-cre) or a dorsal telencephalon enriched cre (Emx1cre/+). Sustained MEK/MAPK activity in the ventral telencephalon (Gsx2e-cre; RosaMEK1DD/+) expanded dorsal lateral ganglionic eminence (dLGE) enriched genes (Gsx2 and Sp8) and oligodendrocyte progenitor cell (OPC) markers (Olig2, Pdgfrα, and Sox10), and also reduced markers in the ventral (v) LGE domain (Isl1 and Foxp1). Activation of MEK/MAPK activity in the dorsal telencephalon (Emx1cre/+; RosaMEK1DD/+) did not initially activate the expression of dLGE or OPC genes at E15.5 but ectopic expression of Gsx2 and OPC markers were observed at E18.5. These results support the idea that MAPK activity as readout by p-ERK1/2 and Etv5 expression is enriched in distinct subdomains of ventral telencephalic progenitors during development. In addition, sustained activation of the MEK/MAPK pathway in the ventral or dorsal telencephalon influences dLGE and OPC identity from progenitors.
Collapse
Affiliation(s)
- Mary Jo Talley
- Graduate Program in Molecular and Developmental Biology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Diana Nardini
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Shenyue Qin
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Carlos E Prada
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Lisa A Ehrman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Ronald R Waclaw
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
29
|
Tiwari D, Schaefer TL, Schroeder-Carter LM, Krzeski JC, Bunk AT, Parkins EV, Snider A, Danzer R, Williams MT, Vorhees CV, Danzer SC, Gross C. The potassium channel Kv4.2 regulates dendritic spine morphology, electroencephalographic characteristics and seizure susceptibility in mice. Exp Neurol 2020; 334:113437. [PMID: 32822706 PMCID: PMC7642025 DOI: 10.1016/j.expneurol.2020.113437] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 01/21/2023]
Abstract
The voltage-gated potassium channel Kv4.2 is a critical regulator of dendritic excitability in the hippocampus and is crucial for dendritic signal integration. Kv4.2 mRNA and protein expression as well as function are reduced in several genetic and pharmacologically induced rodent models of epilepsy and autism. It is not known, however, whether reduced Kv4.2 is just an epiphenomenon or a disease-contributing cause of neuronal hyperexcitability and behavioral impairments in these neurological disorders. To address this question, we used male and female mice heterozygous for a Kv.2 deletion and adult-onset manipulation of hippocampal Kv4.2 expression in male mice to assess the role of Kv4.2 in regulating neuronal network excitability, morphology and anxiety-related behaviors. We observed a reduction in dendritic spine density and reduced proportions of thin and stubby spines but no changes in anxiety, overall activity, or retention of conditioned freezing memory in Kv4.2 heterozygous mice compared with wildtype littermates. Using EEG analyses, we showed elevated theta power and increased spike frequency in Kv4.2 heterozygous mice under basal conditions. In addition, the latency to onset of kainic acid-induced seizures was significantly shortened in Kv4.2 heterozygous mice compared with wildtype littermates, which was accompanied by a significant increase in theta power. By contrast, overexpressing Kv4.2 in wildtype mice through intrahippocampal injection of Kv4.2-expressing lentivirus delayed seizure onset and reduced EEG power. These results suggest that Kv4.2 is an important regulator of neuronal network excitability and dendritic spine morphology, but not anxiety-related behaviors. In the future, manipulation of Kv4.2 expression could be used to alter seizure susceptibility in epilepsy.
Collapse
Affiliation(s)
- Durgesh Tiwari
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Tori L Schaefer
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | - Joseph C Krzeski
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Alexander T Bunk
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Emma V Parkins
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Andrew Snider
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Reese Danzer
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael T Williams
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Charles V Vorhees
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Steve C Danzer
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
30
|
Kuerbitz J, Madhavan M, Ehrman LA, Kohli V, Waclaw RR, Campbell K. Temporally Distinct Roles for the Zinc Finger Transcription Factor Sp8 in the Generation and Migration of Dorsal Lateral Ganglionic Eminence (dLGE)-Derived Neuronal Subtypes in the Mouse. Cereb Cortex 2020; 31:1744-1762. [PMID: 33230547 DOI: 10.1093/cercor/bhaa323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/07/2020] [Accepted: 10/07/2020] [Indexed: 12/29/2022] Open
Abstract
Progenitors in the dorsal lateral ganglionic eminence (dLGE) are known to give rise to olfactory bulb (OB) interneurons and intercalated cells (ITCs) of the amygdala. The dLGE enriched transcription factor Sp8 is required for the normal generation of ITCs as well as OB interneurons, particularly the calretinin (CR)-expressing subtype. In this study, we used a genetic gain-of-function approach in mice to examine the roles Sp8 plays in controlling the development of dLGE-derived neuronal subtypes. Misexpression of Sp8 throughout the ventral telencephalic subventricular zone (SVZ) from early embryonic stages, led to an increased generation of ITCs which was dependent on Tshz1 gene dosage. Additionally, Sp8 misexpression impaired rostral migration of OB interneurons with clusters of CR interneurons seen in the SVZ along with decreased differentiation of calbindin OB interneurons. Sp8 misexpression throughout the ventral telencephalon also reduced ventral LGE neuronal subtypes including striatal projection neurons. Delaying Sp8 misexpression until E14-15 rescued the striatal and amygdala phenotypes but only partially rescued OB interneuron reductions, consistent with an early window of striatal and amygdala neurogenesis and ongoing OB interneuron generation at this late stage. Our results demonstrate critical roles for the timing and neuronal cell-type specificity of Sp8 expression in mouse LGE neurogenesis.
Collapse
Affiliation(s)
- J Kuerbitz
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Medical-Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - M Madhavan
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - L A Ehrman
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Divisions of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - V Kohli
- Divisions of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - R R Waclaw
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Divisions of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - K Campbell
- Divisions of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Divisions of Neurosurgery, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
31
|
Désaubry L, Kanthasamy AG, Nebigil CG. Prokineticin signaling in heart-brain developmental axis: Therapeutic options for heart and brain injuries. Pharmacol Res 2020; 160:105190. [PMID: 32937177 PMCID: PMC7674124 DOI: 10.1016/j.phrs.2020.105190] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/27/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023]
Abstract
Heart and brain development occur simultaneously during the embryogenesis, and both organ development and injuries are interconnected. Early neuronal and cardiac injuries share mutual cellular events, such as angiogenesis and plasticity that could either delay disease progression or, in the long run, result in detrimental health effects. For this reason, the common mechanisms provide a new and previously undervalued window of opportunity for intervention. Because angiogenesis, cardiogenesis and neurogenesis are essential for the development and regeneration of the heart and brain, we discuss therein the role of prokineticin as an angiogenic neuropeptide in heart-brain development and injuries. We focus on the role of prokineticin signaling and the effect of drugs targeting prokineticin receptors in neuroprotection and cardioprotection, with a special emphasis on heart failure, neurodegenerativParkinson's disease and ischemic heart and brain injuries. Indeed, prokineticin triggers common pro-survival signaling pathway in heart and brain. Our review aims at stimulating researchers and clinicians in neurocardiology to focus on the role of prokineticin signaling in the reciprocal interaction between heart and brain. We hope to facilitate the discovery of new treatment strategies, acting in both heart and brain degenerative diseases.
Collapse
Affiliation(s)
- Laurent Désaubry
- Regenerative Nanomedicine, UMR 1260, INSERM, University of Strasbourg, Strasbourg, France
| | - Anumantha G Kanthasamy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Canan G Nebigil
- Regenerative Nanomedicine, UMR 1260, INSERM, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
32
|
Xiao X, Deng H, Furlan A, Yang T, Zhang X, Hwang GR, Tucciarone J, Wu P, He M, Palaniswamy R, Ramakrishnan C, Ritola K, Hantman A, Deisseroth K, Osten P, Huang ZJ, Li B. A Genetically Defined Compartmentalized Striatal Direct Pathway for Negative Reinforcement. Cell 2020; 183:211-227.e20. [PMID: 32937106 DOI: 10.1016/j.cell.2020.08.032] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/02/2020] [Accepted: 08/17/2020] [Indexed: 12/31/2022]
Abstract
The striosome compartment within the dorsal striatum has been implicated in reinforcement learning and regulation of motivation, but how striosomal neurons contribute to these functions remains elusive. Here, we show that a genetically identified striosomal population, which expresses the Teashirt family zinc finger 1 (Tshz1) and belongs to the direct pathway, drives negative reinforcement and is essential for aversive learning in mice. Contrasting a "conventional" striosomal direct pathway, the Tshz1 neurons cause aversion, movement suppression, and negative reinforcement once activated, and they receive a distinct set of synaptic inputs. These neurons are predominantly excited by punishment rather than reward and represent the anticipation of punishment or the motivation for avoidance. Furthermore, inhibiting these neurons impairs punishment-based learning without affecting reward learning or movement. These results establish a major role of striosomal neurons in behaviors reinforced by punishment and moreover uncover functions of the direct pathway unaccounted for in classic models.
Collapse
Affiliation(s)
- Xiong Xiao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Hanfei Deng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Tao Yang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Xian Zhang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Ga-Ram Hwang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Jason Tucciarone
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Priscilla Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Miao He
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | | | - Charu Ramakrishnan
- Howard Hughes Medical Institute (HHMI), Stanford University, Stanford, CA, USA; Department of Bioengineering and Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | | | - Adam Hantman
- HHMI Janelia Research Campus, Ashburn, VA 20147, USA
| | - Karl Deisseroth
- Howard Hughes Medical Institute (HHMI), Stanford University, Stanford, CA, USA; Department of Bioengineering and Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Pavel Osten
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Z Josh Huang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Bo Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
33
|
Co M, Anderson AG, Konopka G. FOXP transcription factors in vertebrate brain development, function, and disorders. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2020; 9:e375. [PMID: 31999079 PMCID: PMC8286808 DOI: 10.1002/wdev.375] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/17/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Abstract
FOXP transcription factors are an evolutionarily ancient protein subfamily coordinating the development of several organ systems in the vertebrate body. Association of their genes with neurodevelopmental disorders has sparked particular interest in their expression patterns and functions in the brain. Here, FOXP1, FOXP2, and FOXP4 are expressed in distinct cell type-specific spatiotemporal patterns in multiple regions, including the cortex, hippocampus, amygdala, basal ganglia, thalamus, and cerebellum. These varied sites and timepoints of expression have complicated efforts to link FOXP1 and FOXP2 mutations to their respective developmental disorders, the former affecting global neural functions and the latter specifically affecting speech and language. However, the use of animal models, particularly those with brain region- and cell type-specific manipulations, has greatly advanced our understanding of how FOXP expression patterns could underlie disorder-related phenotypes. While many questions remain regarding FOXP expression and function in the brain, studies to date have illuminated the roles of these transcription factors in vertebrate brain development and have greatly informed our understanding of human development and disorders. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Marissa Co
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
| | - Ashley G Anderson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Genevieve Konopka
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
34
|
den Hoed J, Fisher SE. Genetic pathways involved in human speech disorders. Curr Opin Genet Dev 2020; 65:103-111. [PMID: 32622339 DOI: 10.1016/j.gde.2020.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022]
Abstract
Rare genetic variants that disrupt speech development provide entry points for deciphering the neurobiological foundations of key human capacities. The value of this approach is illustrated by FOXP2, a transcription factor gene that was implicated in speech apraxia, and subsequently investigated using human cell-based systems and animal models. Advances in next-generation sequencing, coupled to de novo paradigms, facilitated discovery of etiological variants in additional genes in speech disorder cohorts. As for other neurodevelopmental syndromes, gene-driven studies show blurring of boundaries between diagnostic categories, with some risk genes shared across speech disorders, intellectual disability and autism. Convergent evidence hints at involvement of regulatory genes co-expressed in early human brain development, suggesting that etiological pathways could be amenable for investigation in emerging neural models such as cerebral organoids.
Collapse
Affiliation(s)
- Joery den Hoed
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands; International Max Planck Research School for Language Sciences, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, The Netherlands.
| |
Collapse
|
35
|
Roychoudhury K, Salomone J, Qin S, Cain B, Adam M, Potter SS, Nakafuku M, Gebelein B, Campbell K. Physical interactions between Gsx2 and Ascl1 balance progenitor expansion versus neurogenesis in the mouse lateral ganglionic eminence. Development 2020; 147:dev.185348. [PMID: 32122989 DOI: 10.1242/dev.185348] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/13/2020] [Indexed: 12/15/2022]
Abstract
The Gsx2 homeodomain transcription factor promotes neural progenitor identity in the lateral ganglionic eminence (LGE), despite upregulating the neurogenic factor Ascl1. How this balance in maturation is maintained is unclear. Here, we show that Gsx2 and Ascl1 are co-expressed in subapical progenitors that have unique transcriptional signatures in LGE ventricular zone (VZ) cells. Moreover, whereas Ascl1 misexpression promotes neurogenesis in dorsal telencephalic progenitors, the co-expression of Gsx2 with Ascl1 inhibits neurogenesis. Using luciferase assays, we found that Gsx2 reduces the ability of Ascl1 to activate gene expression in a dose-dependent and DNA binding-independent manner. Furthermore, Gsx2 physically interacts with the basic helix-loop-helix (bHLH) domain of Ascl1, and DNA-binding assays demonstrated that this interaction interferes with the ability of Ascl1 to bind DNA. Finally, we modified a proximity ligation assay for tissue sections and found that Ascl1-Gsx2 interactions are enriched within LGE VZ progenitors, whereas Ascl1-Tcf3 (E-protein) interactions predominate in the subventricular zone. Thus, Gsx2 contributes to the balance between progenitor maintenance and neurogenesis by physically interacting with Ascl1, interfering with its DNA binding and limiting neurogenesis within LGE progenitors.
Collapse
Affiliation(s)
- Kaushik Roychoudhury
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Joseph Salomone
- Graduate Program in Molecular and Developmental Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA.,Medical-Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Shenyue Qin
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Brittany Cain
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Masato Nakafuku
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Brian Gebelein
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Kenneth Campbell
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
36
|
Beyeler A, Dabrowska J. Neuronal diversity of the amygdala and the bed nucleus of the stria terminalis. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2020; 26:63-100. [PMID: 32792868 DOI: 10.1016/b978-0-12-815134-1.00003-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Anna Beyeler
- Neurocentre Magendie, French National Institutes of Health (INSERM) unit 1215, Neurocampus of Bordeaux University, Bordeaux, France
| | - Joanna Dabrowska
- Center for the Neurobiology of Stress Resilience and Psychiatric Disorders, Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
37
|
Zoubovsky SP, Hoseus S, Tumukuntala S, Schulkin JO, Williams MT, Vorhees CV, Muglia LJ. Chronic psychosocial stress during pregnancy affects maternal behavior and neuroendocrine function and modulates hypothalamic CRH and nuclear steroid receptor expression. Transl Psychiatry 2020; 10:6. [PMID: 32066677 PMCID: PMC7026416 DOI: 10.1038/s41398-020-0704-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/09/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023] Open
Abstract
Postpartum depression (PPD) affects up to 20% of mothers and has negative consequences for both mother and child. Although exposure to psychosocial stress during pregnancy and abnormalities in the hypothalamic pituitary adrenal (HPA) axis have been linked to PPD, molecular changes in the brain that contribute to this disease remain unknown. This study utilized a novel chronic psychosocial stress paradigm during pregnancy (CGS) to investigate the effects of psychosocial stress on maternal behavior, neuroendocrine function, and gene expression changes in molecular regulators of the HPA axis in the early postpartum period. Postpartum female mice exposed to CGS display abnormalities in maternal behavior, including fragmented and erratic maternal care patterns, and the emergence of depression and anxiety-like phenotypes. Dysregulation in postpartum HPA axis function, evidenced by blunted circadian peak and elevation of stress-induced corticosterone levels, was accompanied by increased CRH mRNA expression and a reduction in CRH receptor 1 in the paraventricular nucleus of the hypothalamus (PVN). We further observed decreased PVN expression of nuclear steroid hormone receptors associated with CRH transcription, suggesting these molecular changes could underlie abnormalities in postpartum HPA axis and behavior observed. Overall, our study demonstrates that psychosocial stress during pregnancy induces changes in neuroendocrine function and maternal behavior in the early postpartum period and introduces our CGS paradigm as a viable model that can be used to further dissect the molecular defects that lead to PPD.
Collapse
Affiliation(s)
- Sandra P Zoubovsky
- Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sarah Hoseus
- Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shivani Tumukuntala
- Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jay O Schulkin
- Department of Neuroscience, Georgetown University, Washington, DC, USA
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Louis J Muglia
- Center for the Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
38
|
Guo T, Liu G, Du H, Wen Y, Wei S, Li Z, Tao G, Shang Z, Song X, Zhang Z, Xu Z, You Y, Chen B, Rubenstein JL, Yang Z. Dlx1/2 are Central and Essential Components in the Transcriptional Code for Generating Olfactory Bulb Interneurons. Cereb Cortex 2019; 29:4831-4849. [PMID: 30796806 PMCID: PMC6917526 DOI: 10.1093/cercor/bhz018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/03/2019] [Accepted: 01/26/2019] [Indexed: 12/22/2022] Open
Abstract
Generation of olfactory bulb (OB) interneurons requires neural stem/progenitor cell specification, proliferation, differentiation, and young interneuron migration and maturation. Here, we show that the homeobox transcription factors Dlx1/2 are central and essential components in the transcriptional code for generating OB interneurons. In Dlx1/2 constitutive null mutants, the differentiation of GSX2+ and ASCL1+ neural stem/progenitor cells in the dorsal lateral ganglionic eminence is blocked, resulting in a failure of OB interneuron generation. In Dlx1/2 conditional mutants (hGFAP-Cre; Dlx1/2F/- mice), GSX2+ and ASCL1+ neural stem/progenitor cells in the postnatal subventricular zone also fail to differentiate into OB interneurons. In contrast, overexpression of Dlx1&2 in embryonic mouse cortex led to ectopic production of OB-like interneurons that expressed Gad1, Sp8, Sp9, Arx, Pbx3, Etv1, Tshz1, and Prokr2. Pax6 mutants generate cortical ectopia with OB-like interneurons, but do not do so in compound Pax6; Dlx1/2 mutants. We propose that DLX1/2 promote OB interneuron development mainly through activating the expression of Sp8/9, which further promote Tshz1 and Prokr2 expression. Based on this study, in combination with earlier ones, we propose a transcriptional network for the process of OB interneuron development.
Collapse
Affiliation(s)
- Teng Guo
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Guoping Liu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Heng Du
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yan Wen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Song Wei
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhenmeiyu Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Guangxu Tao
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zicong Shang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Xiaolei Song
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhejun Xu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yan You
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Bin Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - John L Rubenstein
- Department of Psychiatry, Nina Ireland Laboratory of Developmental Neurobiology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| |
Collapse
|
39
|
Chaimowicz C, Ruffault PL, Chéret C, Woehler A, Zampieri N, Fortin G, Garratt AN, Birchmeier C. Teashirt 1 (Tshz1) is essential for the development, survival and function of hypoglossal and phrenic motor neurons in mouse. Development 2019; 146:dev.174045. [PMID: 31427287 PMCID: PMC6765129 DOI: 10.1242/dev.174045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 08/09/2019] [Indexed: 11/20/2022]
Abstract
Feeding and breathing are essential motor functions and rely on the activity of hypoglossal and phrenic motor neurons that innervate the tongue and diaphragm, respectively. Little is known about the genetic programs that control the development of these neuronal subtypes. The transcription factor Tshz1 is strongly and persistently expressed in developing hypoglossal and phrenic motor neurons. We used conditional mutation of Tshz1 in the progenitor zone of motor neurons (Tshz1MN Δ) to show that Tshz1 is essential for survival and function of hypoglossal and phrenic motor neurons. Hypoglossal and phrenic motor neurons are born in correct numbers, but many die between embryonic day 13.5 and 14.5 in Tshz1MN Δ mutant mice. In addition, innervation and electrophysiological properties of phrenic and hypoglossal motor neurons are altered. Severe feeding and breathing problems accompany this developmental deficit. Although motor neuron survival can be rescued by elimination of the pro-apoptotic factor Bax, innervation, feeding and breathing defects persist in Bax-/-; Tshz1MN Δ mutants. We conclude that Tshz1 is an essential transcription factor for the development and physiological function of phrenic and hypoglossal motor neurons.
Collapse
Affiliation(s)
- Charlotte Chaimowicz
- Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Society, 13125 Berlin, Germany
| | - Pierre-Louis Ruffault
- Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Society, 13125 Berlin, Germany
| | - Cyril Chéret
- Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Society, 13125 Berlin, Germany
| | - Andrew Woehler
- Systems Biology Imaging, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Society, 13125 Berlin, Germany
| | - Niccolò Zampieri
- Development and Function of Neural Circuits, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Society, 13125 Berlin, Germany
| | - Gilles Fortin
- UMR9197, CNRS/Université Paris-Sud, Paris-Saclay Institute of Neuroscience, 1 Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Alistair N Garratt
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Virchowweg 6, 10117 Berlin, Germany
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Society, 13125 Berlin, Germany
| |
Collapse
|
40
|
Abstract
Nobel laureate Nikolaas Tinbergen provided clear criteria for declaring a neuroscience problem solved, criteria which despite the passage of more than 50 years and vastly expanded neuroscience tool kits remain applicable today. Tinbergen said for neuroscientists to claim that a behavior is understood, they must correspondingly understand its (i) development and its (ii) mechanisms and its (iii) function and its (iv) evolution. Now, all four of these domains represent hotbeds of current experimental work, each using arrays of new techniques which overlap only partly. Thus, as new methodologies come online, from single-nerve-cell RNA sequencing, for example, to smart FISH, large-scale calcium imaging from cortex and deep brain structures, computational ethology, and so on, one person, however smart, cannot master everything. Our response to the likely “fracturing” of neuroscience recognizes the value of ever larger consortia. This response suggests new kinds of problems for (i) funding and (ii) the fair distribution of credit, especially for younger scientists.
Collapse
|
41
|
Abstract
The neural mechanisms underlying emotional valence are at the interface between perception and action, integrating inputs from the external environment with past experiences to guide the behavior of an organism. Depending on the positive or negative valence assigned to an environmental stimulus, the organism will approach or avoid the source of the stimulus. Multiple convergent studies have demonstrated that the amygdala complex is a critical node of the circuits assigning valence. Here we examine the current progress in identifying valence coding properties of neural populations in different nuclei of the amygdala, based on their activity, connectivity, and gene expression profile.
Collapse
Affiliation(s)
- Michele Pignatelli
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, 02139 MA, USA
| | - Anna Beyeler
- Neurocentre Magendie, INSERM 1215, Université de Bordeaux, 146 Rue Léo Saignat, 33000 Bordeaux, France
| |
Collapse
|