1
|
Deore R, Ansari R, Awathale SN, Shelke M, Badwaik HR, Goyal SN, Nakhate KT. Lycopene alleviates BCG-induced depressive phenotypes in mice by disrupting 5-HT3 receptor - IDO1 interplay in the brain. Eur J Pharmacol 2024; 977:176707. [PMID: 38830456 DOI: 10.1016/j.ejphar.2024.176707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/20/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
The 5-HT3 receptor and indoleamine 2,3-dioxygenase 1 (IDO1) enzyme play a crucial role in the pathogenesis of depression as their activation reduces serotonin contents in the brain. Since molecular docking analysis revealed lycopene as a potent 5-HT3 receptor antagonist and IDO1 inhibitor, we hypothesized that lycopene might disrupt the interplay between the 5-HT3 receptor and IDO1 to mitigate depression. In mice, the depression-like phenotypes were induced by inoculating Bacillus Calmette-Guerin (BCG). Lycopene (intraperitoneal; i.p.) was administered alone or in combination with 5-HT3 receptor antagonist ondansetron (i.p.) or IDO1 inhibitor minocycline (i.p.), and the behavioral screening was performed by the sucrose preference test, open field test, tail suspension test, and splash test which are based on the different principles. Further, the brains were subjected to the biochemical analysis of serotonin and its precursor tryptophan by the HPLC. The results showed depression-like behavior in BCG-inoculated mice, which was reversed by lycopene administration. Moreover, prior treatment with ondansetron or minocycline potentiated the antidepressant action of lycopene. Minocycline pretreatment also enhanced the antidepressant effect of ondansetron indicating the regulation of IDO1 activity by 5-HT3 receptor-triggered signaling. Biochemical analysis of brain samples revealed a drastic reduction in the levels of tryptophan and serotonin in depressed animals, which were restored following treatment with lycopene and its combination with ondansetron or minocycline. Taken together, the data from molecular docking, behavioral experiments, and biochemical estimation suggest that lycopene might block the 5-HT3 receptor and consequently inhibit the activity of IDO1 to ameliorate BCG-induced depression in mice.
Collapse
Affiliation(s)
- Rucha Deore
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Rashid Ansari
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Sanjay N Awathale
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Madhav Shelke
- Department of Quality Assurance, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Hemant R Badwaik
- Department of Pharmaceutical Chemistry, Shri Shankaracharya Institute of Pharmaceutical Sciences and Research, Bhilai, 490020, Chhattisgarh, India
| | - Sameer N Goyal
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Kartik T Nakhate
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India.
| |
Collapse
|
2
|
Machold R, Rudy B. Genetic approaches to elucidating cortical and hippocampal GABAergic interneuron diversity. Front Cell Neurosci 2024; 18:1414955. [PMID: 39113758 PMCID: PMC11303334 DOI: 10.3389/fncel.2024.1414955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
GABAergic interneurons (INs) in the mammalian forebrain represent a diverse population of cells that provide specialized forms of local inhibition to regulate neural circuit activity. Over the last few decades, the development of a palette of genetic tools along with the generation of single-cell transcriptomic data has begun to reveal the molecular basis of IN diversity, thereby providing deep insights into how different IN subtypes function in the forebrain. In this review, we outline the emerging picture of cortical and hippocampal IN speciation as defined by transcriptomics and developmental origin and summarize the genetic strategies that have been utilized to target specific IN subtypes, along with the technical considerations inherent to each approach. Collectively, these methods have greatly facilitated our understanding of how IN subtypes regulate forebrain circuitry via cell type and compartment-specific inhibition and thus have illuminated a path toward potential therapeutic interventions for a variety of neurocognitive disorders.
Collapse
Affiliation(s)
- Robert Machold
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Bernardo Rudy
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
3
|
Machold R, Dellal S, Valero M, Zurita H, Kruglikov I, Meng JH, Hanson JL, Hashikawa Y, Schuman B, Buzsáki G, Rudy B. Id2 GABAergic interneurons comprise a neglected fourth major group of cortical inhibitory cells. eLife 2023; 12:e85893. [PMID: 37665123 PMCID: PMC10581691 DOI: 10.7554/elife.85893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 08/21/2023] [Indexed: 09/05/2023] Open
Abstract
Cortical GABAergic interneurons (INs) represent a diverse population of mainly locally projecting cells that provide specialized forms of inhibition to pyramidal neurons and other INs. Most recent work on INs has focused on subtypes distinguished by expression of Parvalbumin (PV), Somatostatin (SST), or Vasoactive Intestinal Peptide (VIP). However, a fourth group that includes neurogliaform cells (NGFCs) has been less well characterized due to a lack of genetic tools. Here, we show that these INs can be accessed experimentally using intersectional genetics with the gene Id2. We find that outside of layer 1 (L1), the majority of Id2 INs are NGFCs that express high levels of neuropeptide Y (NPY) and exhibit a late-spiking firing pattern, with extensive local connectivity. While much sparser, non-NGFC Id2 INs had more variable properties, with most cells corresponding to a diverse group of INs that strongly expresses the neuropeptide CCK. In vivo, using silicon probe recordings, we observed several distinguishing aspects of NGFC activity, including a strong rebound in activity immediately following the cortical down state during NREM sleep. Our study provides insights into IN diversity and NGFC distribution and properties, and outlines an intersectional genetics approach for further study of this underappreciated group of INs.
Collapse
Affiliation(s)
- Robert Machold
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Shlomo Dellal
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Manuel Valero
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Hector Zurita
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Ilya Kruglikov
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - John Hongyu Meng
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Center for Neural Science, New York UniversityNew YorkUnited States
| | - Jessica L Hanson
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Yoshiko Hashikawa
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Benjamin Schuman
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - György Buzsáki
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
| | - Bernardo Rudy
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
4
|
Cao JW, Mao XY, Zhu L, Zhou ZS, Jiang SN, Liu LY, Zhang SQ, Fu Y, Xu WD, Yu YC. Correlation Analysis of Molecularly-Defined Cortical Interneuron Populations with Morpho-Electric Properties in Layer V of Mouse Neocortex. Neurosci Bull 2023; 39:1069-1086. [PMID: 36422797 PMCID: PMC10313633 DOI: 10.1007/s12264-022-00983-x] [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: 06/16/2022] [Accepted: 08/16/2022] [Indexed: 11/25/2022] Open
Abstract
Cortical interneurons can be categorized into distinct populations based on multiple modalities, including molecular signatures and morpho-electrical (M/E) properties. Recently, many transcriptomic signatures based on single-cell RNA-seq have been identified in cortical interneurons. However, whether different interneuron populations defined by transcriptomic signature expressions correspond to distinct M/E subtypes is still unknown. Here, we applied the Patch-PCR approach to simultaneously obtain the M/E properties and messenger RNA (mRNA) expression of >600 interneurons in layer V of the mouse somatosensory cortex (S1). Subsequently, we identified 11 M/E subtypes, 9 neurochemical cell populations (NCs), and 20 transcriptomic cell populations (TCs) in this cortical lamina. Further analysis revealed that cells in many NCs and TCs comprised several M/E types and were difficult to clearly distinguish morpho-electrically. A similar analysis of layer V interneurons of mouse primary visual cortex (V1) and motor cortex (M1) gave results largely comparable to S1. Comparison between S1, V1, and M1 suggested that, compared to V1, S1 interneurons were morpho-electrically more similar to M1. Our study reveals the presence of substantial M/E variations in cortical interneuron populations defined by molecular expression.
Collapse
Affiliation(s)
- Jun-Wei Cao
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Xiao-Yi Mao
- School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Liang Zhu
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Zhi-Shuo Zhou
- School of Data Science, Fudan University, Shanghai, 200433, China
| | - Shao-Na Jiang
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Lin-Yun Liu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Shu-Qing Zhang
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yinghui Fu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Wen-Dong Xu
- The National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yong-Chun Yu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
Fossati G, Kiss-Bodolay D, Prados J, Chéreau R, Husi E, Cadilhac C, Gomez L, Silva BA, Dayer A, Holtmaat A. Bimodal modulation of L1 interneuron activity in anterior cingulate cortex during fear conditioning. Front Neural Circuits 2023; 17:1138358. [PMID: 37334059 PMCID: PMC10272719 DOI: 10.3389/fncir.2023.1138358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
The anterior cingulate cortex (ACC) plays a crucial role in encoding, consolidating and retrieving memories related to emotionally salient experiences, such as aversive and rewarding events. Various studies have highlighted its importance for fear memory processing, but its circuit mechanisms are still poorly understood. Cortical layer 1 (L1) of the ACC might be a particularly important site of signal integration, since it is a major entry point for long-range inputs, which is tightly controlled by local inhibition. Many L1 interneurons express the ionotropic serotonin receptor 3a (5HT3aR), which has been implicated in post-traumatic stress disorder and in models of anxiety. Hence, unraveling the response dynamics of L1 interneurons and subtypes thereof during fear memory processing may provide important insights into the microcircuit organization regulating this process. Here, using 2-photon laser scanning microscopy of genetically encoded calcium indicators through microprisms in awake mice, we longitudinally monitored over days the activity of L1 interneurons in the ACC in a tone-cued fear conditioning paradigm. We observed that tones elicited responses in a substantial fraction of the imaged neurons, which were significantly modulated in a bidirectional manner after the tone was associated to an aversive stimulus. A subpopulation of these neurons, the neurogliaform cells (NGCs), displayed a net increase in tone-evoked responses following fear conditioning. Together, these results suggest that different subpopulations of L1 interneurons may exert distinct functions in the ACC circuitry regulating fear learning and memory.
Collapse
Affiliation(s)
- Giuliana Fossati
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Neuro Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Daniel Kiss-Bodolay
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Neurosurgery, Geneva University Hospitals, Geneva, Switzerland
- Lemanic Neuroscience Doctoral School, University of Geneva, Geneva, Switzerland
| | - Julien Prados
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Ronan Chéreau
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Elodie Husi
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christelle Cadilhac
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Lucia Gomez
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Bianca A. Silva
- Neuro Center, IRCCS Humanitas Research Hospital, Milan, Italy
- National Research Council of Italy, Institute of Neuroscience, Milan, Italy
| | - Alexandre Dayer
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Anthony Holtmaat
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
6
|
Li J, Zheng S, Dong Y, Xu H, Zhu Y, Weng J, Sun D, Wang S, Xiao L, Jiang Y. Histone Methyltransferase SETDB1 Regulates the Development of Cortical Htr3a-Positive Interneurons and Mood Behaviors. Biol Psychiatry 2023; 93:279-290. [PMID: 36335068 DOI: 10.1016/j.biopsych.2022.08.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 08/02/2022] [Accepted: 08/22/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND GABAergic (gamma-aminobutyric acidergic) interneurons (INs) are highly heterogeneous, and Htr3a labels a subpopulation of cortical INs originating from the embryonic caudal ganglionic eminence. SETDB1 is one of the histone H3K9 methyltransferases and plays an essential role in the excitatory neurons, but its role in regulating cortical inhibitory INs remains largely unknown. METHODS In this study, we generated transgenic mice with conditional knockout of Setdb1 in neural progenitor cells (Setdb1-NS-cKO) and GABAergic neurons (Setdb1-Gad2-cKO). In addition, we performed RNA sequencing, ATAC-seq (assay for transposase-accessible chromatin with sequencing), chromatin immunoprecipitation sequencing, luciferase assay, chromatin conformation capture, and CRISPR (clustered regularly interspaced short palindromic repeats)/dCas9 to study the epigenetic mechanism underlying SETDB1-mediated transcriptional regulation of Htr3a. We also performed in situ hybridization and whole-cell recording to evaluate the functional properties of cortical Htr3a+ INs and behavioral tests for mood. RESULTS We detected significant upregulation of Htr3a expression in the embryonic ganglionic eminence of Setdb1-NS-cKO and identified the endogenous retroviral sequence RMER21B as a new target of SETDB1. RMER21B showed enhancer activity and formed distal chromatin interaction with the promoter of Htr3a. In addition, we observed an increased number and enhanced excitability of Htr3a+ INs in the knockout cortex. Moreover, Setdb1-Gad2-cKO mice exhibited anxiety- and depressive-like behaviors, which were partially reversed by a 5-HT3 receptor antagonist. CONCLUSIONS These findings suggest that SETDB1 represses Htr3a transcription via RMER21B-mediated distal chromatin interaction in the embryonic ganglionic eminence and regulates the development of cortical Htr3a+ INs and mood behaviors.
Collapse
Affiliation(s)
- Jiaqi Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shenghui Zheng
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yuhao Dong
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Hao Xu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yueyan Zhu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jie Weng
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Daijing Sun
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | | | - Lei Xiao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yan Jiang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Bravo K, González-Ortiz M, Beltrán-Castillo S, Cáceres D, Eugenín J. Development of the Placenta and Brain Are Affected by Selective Serotonin Reuptake Inhibitor Exposure During Critical Periods. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:179-198. [PMID: 37466774 DOI: 10.1007/978-3-031-32554-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are usually prescribed to treat major depression and anxiety disorders. Fetal brain development exhibits dependency on serotonin (5-hydroxytryptamine, 5-HT) from maternal, placental, and fetal brain sources. At very early fetal stages, fetal serotonin is provided by maternal and placental sources. However, in later fetal stages, brain sources are indispensable for the appropriate development of neural circuitry and the rise of emergent functions implied in behavior acquisition. Thus, susceptible serotonin-related critical periods are recognized, involving the early maternal and placental 5-HT synthesis and the later endogenous 5-HT synthesis in the fetal brain. Acute and chronic exposure to SSRIs during these critical periods may result in short- and long-term placental and brain dysfunctions affecting intrauterine and postnatal life. Maternal and fetal cells express serotonin receptors which make them susceptible to changes in serotonin levels influenced by SSRIs. SSRIs block the serotonin transporter (SERT), which is required for 5-HT reuptake from the synaptic cleft into the presynaptic neuron. Chronic SSRI administration leads to pre- and postsynaptic 5-HT receptor rearrangement. In this review, we focus on the effects of SSRIs administered during critical periods upon placentation and brain development to be considered in evaluating the risk-safety balance in the clinical use of SSRIs.
Collapse
Affiliation(s)
- Karina Bravo
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile USACH, Santiago, Chile.
- Facultad de Ingeniería, Universidad Autónoma de Chile, Santiago, Chile.
| | - Marcelo González-Ortiz
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
| | - Sebastian Beltrán-Castillo
- Centro integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Daniela Cáceres
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile USACH, Santiago, Chile
| | - Jaime Eugenín
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile USACH, Santiago, Chile
| |
Collapse
|
8
|
Venkataramanappa S, Saaber F, Abe P, Schütz D, Kumar PA, Stumm R. Cxcr4 and Ackr3 regulate allocation of caudal ganglionic eminence-derived interneurons to superficial cortical layers. Cell Rep 2022; 40:111157. [PMID: 35926459 DOI: 10.1016/j.celrep.2022.111157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/17/2022] [Accepted: 07/13/2022] [Indexed: 11/29/2022] Open
Abstract
The function of the cerebral cortex depends on various types of interneurons (cortical interneurons [cINs]) and their appropriate allocation to the cortical layers. Caudal ganglionic eminence-derived cINs (cGE-cINs) are enriched in superficial layers. Developmental mechanisms directing cGE-cINs toward superficial layers remain poorly understood. We examine how developmental and final positioning of cGE-cINs are influenced by the Cxcl12, Cxcr4, Ackr3 module, the chief attractant system guiding medial ganglionic eminence-derived cINs (mGE-cINs). We find that Cxcl12 attracts cGE-cINs through Cxcr4 and supports their layer-specific positioning in the developing cortex. This requires the prevention of excessive Cxcr4 stimulation by Ackr3-mediated Cxcl12 sequestration. Postnatally, Ackr3 confines Cxcl12 action to the marginal zone. Unlike mGE-cINs, cGE-cINs continue to express Cxcr4 at early postnatal stages, which permits cGE-cINs to become positioned in the forming layer 1. Thus, chemoattraction by Cxcl12 guides cGE-cINs and holds them in superficial cortical layers.
Collapse
Affiliation(s)
| | - Friederike Saaber
- Institute of Pharmacology and Toxicology, University Hospital Jena, Jena, Germany
| | - Philipp Abe
- Institute of Pharmacology and Toxicology, University Hospital Jena, Jena, Germany
| | - Dagmar Schütz
- Institute of Pharmacology and Toxicology, University Hospital Jena, Jena, Germany
| | - Praveen Ashok Kumar
- Institute of Pharmacology and Toxicology, University Hospital Jena, Jena, Germany
| | - Ralf Stumm
- Institute of Pharmacology and Toxicology, University Hospital Jena, Jena, Germany.
| |
Collapse
|
9
|
Perrenoud Q, Leclerc C, Geoffroy H, Vitalis T, Richetin K, Rampon C, Gallopin T. Molecular and electrophysiological features of GABAergic neurons in the dentate gyrus reveal limited homology with cortical interneurons. PLoS One 2022; 17:e0270981. [PMID: 35802727 PMCID: PMC9269967 DOI: 10.1371/journal.pone.0270981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
GABAergic interneurons tend to diversify into similar classes across telencephalic regions. However, it remains unclear whether the electrophysiological and molecular properties commonly used to define these classes are discriminant in the hilus of the dentate gyrus. Here, using patch-clamp combined with single cell RT-PCR, we compare the relevance of commonly used electrophysiological and molecular features for the clustering of GABAergic interneurons sampled from the mouse hilus and primary sensory cortex. While unsupervised clustering groups cortical interneurons into well-established classes, it fails to provide a convincing partition of hilar interneurons. Statistical analysis based on resampling indicates that hilar and cortical GABAergic interneurons share limited homology. While our results do not invalidate the use of classical molecular marker in the hilus, they indicate that classes of hilar interneurons defined by the expression of molecular markers do not exhibit strongly discriminating electrophysiological properties.
Collapse
Affiliation(s)
- Quentin Perrenoud
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Clémence Leclerc
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Hélène Geoffroy
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Tania Vitalis
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Kevin Richetin
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Thierry Gallopin
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
- * E-mail:
| |
Collapse
|
10
|
Llorca A, Deogracias R. Origin, Development, and Synaptogenesis of Cortical Interneurons. Front Neurosci 2022; 16:929469. [PMID: 35833090 PMCID: PMC9272671 DOI: 10.3389/fnins.2022.929469] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral cortex represents one of the most recent and astonishing inventions of nature, responsible of a large diversity of functions that range from sensory processing to high-order cognitive abilities, such as logical reasoning or language. Decades of dedicated study have contributed to our current understanding of this structure, both at structural and functional levels. A key feature of the neocortex is its outstanding richness in cell diversity, composed by multiple types of long-range projecting neurons and locally connecting interneurons. In this review, we will describe the great diversity of interneurons that constitute local neocortical circuits and summarize the mechanisms underlying their development and their assembly into functional networks.
Collapse
Affiliation(s)
- Alfredo Llorca
- Visual Neuroscience Laboratory, Centre for Discovery Brain Sciences, School of Biomedical Sciences, University of Edinburgh, Edinburg, United Kingdom
- *Correspondence: Alfredo Llorca
| | - Ruben Deogracias
- Neuronal Circuits Formation and Brain Disorders Laboratory, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Department of Cell Biology and Pathology, School of Medicine, University of Salamanca, Salamanca, Spain
- Ruben Deogracias
| |
Collapse
|
11
|
Wu J, Zhao Z, Shi Y, He M. Cortical VIP + Interneurons in the Upper and Deeper Layers Are Transcriptionally Distinct. J Mol Neurosci 2022; 72:1779-1795. [PMID: 35708842 DOI: 10.1007/s12031-022-02040-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/07/2022] [Indexed: 12/19/2022]
Abstract
Different interneuron classes have distinct laminar distribution patterns which contribute to the layer-specific organization of cortical microcircuits. However, laminar differences within the same interneuron classes are not well recognized. Despite systematic efforts towards neuron cell-type taxonomy in the neocortex by single-cell transcriptomics, less attention has been driven towards laminar differences in interneurons compared to projection neurons. VIP+ interneurons are the major interneuron class that mostly populate superficial layers and mediate disinhibition. A few reports noted the morphological and electrophysiological differences between VIP+ interneurons residing in layers I-III (upper layer) and layers IV-VI (deeper layer), but little is known about their molecular differences. Here, we delineated the laminar difference in their transcriptome employing single-cell RNA sequencing (scRNAseq) data from public databases. Analysis of 1175 high-quality VIP+ interneurons in the primary visual cortex (VISp) showed that the upper layer and deeper layer VIP+ interneurons are transcriptionally distinct distinguished by genes implicated in synapse organization and regulation of membrane potential. Similar differences are also observed in the anterior lateral motor cortex (ALM) and primary motor cortex (MOp). Cross-comparing between the top 10 differentially expressed genes (DEGs) with Allen Mouse Brain in situ hybridization database, we identified Tac2 and CxCl14 as potential marker genes of upper layer VIP+ interneurons across most cortical regions. Importantly, such expression patterns are conserved in the human brain. Together, we revealed significant laminar differences in transcriptomic profiles within VIP+ interneurons, which provided new insight into their molecular heterogeneity that may contribute to their functional diversity.
Collapse
Affiliation(s)
- Jinyun Wu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhirong Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yun Shi
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Miao He
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Tian D, Izumi SI. Transcranial Magnetic Stimulation and Neocortical Neurons: The Micro-Macro Connection. Front Neurosci 2022; 16:866245. [PMID: 35495053 PMCID: PMC9039343 DOI: 10.3389/fnins.2022.866245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/28/2022] [Indexed: 12/20/2022] Open
Abstract
Understanding the operation of cortical circuits is an important and necessary task in both neuroscience and neurorehabilitation. The functioning of the neocortex results from integrative neuronal activity, which can be probed non-invasively by transcranial magnetic stimulation (TMS). Despite a clear indication of the direct involvement of cortical neurons in TMS, no explicit connection model has been made between the microscopic neuronal landscape and the macroscopic TMS outcome. Here we have performed an integrative review of multidisciplinary evidence regarding motor cortex neurocytology and TMS-related neurophysiology with the aim of elucidating the micro–macro connections underlying TMS. Neurocytological evidence from animal and human studies has been reviewed to describe the landscape of the cortical neurons covering the taxonomy, morphology, circuit wiring, and excitatory–inhibitory balance. Evidence from TMS studies in healthy humans is discussed, with emphasis on the TMS pulse and paradigm selectivity that reflect the underlying neural circuitry constitution. As a result, we propose a preliminary neuronal model of the human motor cortex and then link the TMS mechanisms with the neuronal model by stimulus intensity, direction of induced current, and paired-pulse timing. As TMS bears great developmental potential for both a probe and modulator of neural network activity and neurotransmission, the connection model will act as a foundation for future combined studies of neurocytology and neurophysiology, as well as the technical advances and application of TMS.
Collapse
Affiliation(s)
- Dongting Tian
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduates School of Medicine, Sendai, Japan
- *Correspondence: Dongting Tian,
| | - Shin-Ichi Izumi
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduates School of Medicine, Sendai, Japan
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
- Shin-Ichi Izumi,
| |
Collapse
|
13
|
Apicella AJ, Marchionni I. VIP-Expressing GABAergic Neurons: Disinhibitory vs. Inhibitory Motif and Its Role in Communication Across Neocortical Areas. Front Cell Neurosci 2022; 16:811484. [PMID: 35221922 PMCID: PMC8867699 DOI: 10.3389/fncel.2022.811484] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
GABAergic neurons play a crucial role in shaping cortical activity. Even though GABAergic neurons constitute a small fraction of cortical neurons, their peculiar morphology and functional properties make them an intriguing and challenging task to study. Here, we review the basic anatomical features, the circuit properties, and the possible role in the relevant behavioral task of a subclass of GABAergic neurons that express vasoactive intestinal polypeptide (VIP). These studies were performed using transgenic mice in which the VIP-expressing neurons can be recognized using fluorescent proteins and optogenetic manipulation to control (or regulate) their electrical activity. Cortical VIP-expressing neurons are more abundant in superficial cortical layers than other cortical layers, where they are mainly studied. Optogenetic and paired recordings performed in ex vivo cortical preparations show that VIP-expressing neurons mainly exert their inhibitory effect onto somatostatin-expressing (SOM) inhibitory neurons, leading to a disinhibitory effect onto excitatory pyramidal neurons. However, this subclass of GABAergic neurons also releases neurotransmitters onto other GABAergic and non-GABAergic neurons, suggesting other possible circuit roles than a disinhibitory effect. The heterogeneity of VIP-expressing neurons also suggests their involvement and recruitment during different functions via the inhibition/disinhibition of GABAergic and non-GABAergic neurons locally and distally, depending on the specific local circuit in which they are embedded, with potential effects on the behavioral states of the animal. Although VIP-expressing neurons represent only a tiny fraction of GABAergic inhibitory neurons in the cortex, these neurons’ selective activation/inactivation could produce a relevant behavioral effect in the animal. Regardless of the increasing finding and discoveries on this subclass of GABAergic neurons, there is still a lot of missing information, and more studies should be done to unveil their role at the circuit and behavior level in different cortical layers and across different neocortical areas.
Collapse
Affiliation(s)
- Alfonso Junior Apicella
- Department of Biology, Neurosciences Institute, University of Texas at San Antonio, San Antonio, TX, United States
| | - Ivan Marchionni
- Department of Biomedical Sciences, University of Padova, Padua, Italy.,Padova Neuroscience Center (PNC), University of Padova, Padua, Italy
| |
Collapse
|
14
|
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: 9] [Impact Index Per Article: 3.0] [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
|
15
|
Lichtensteiger W, Bassetti-Gaille C, Rehrauer H, Georgijevic JK, Tresguerres JAF, Schlumpf M. Converging Effects of Three Different Endocrine Disrupters on Sox and Pou Gene Expression in Developing Rat Hippocampus: Possible Role of microRNA in Sex Differences. Front Genet 2021; 12:718796. [PMID: 34858468 PMCID: PMC8632217 DOI: 10.3389/fgene.2021.718796] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/20/2021] [Indexed: 11/22/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs) can impair hippocampus-dependent behaviors in rat offspring and in children. In search for key processes underlying this effect, we compared the transcriptomes of rat hippocampus on postnatal day 6 after gestational and lactational exposure to three different EDCs at doses known to impair development of learning and memory. Aroclor 1254, a commercial PCB mixture (5 mg/kg or 0.5 mg/kg), or bisphenol A (5 mg/kg or 0.5 mg/kg) were administered in chow, chlorpyrifos (3 mg/kg or 1 mg/kg) was injected subcutaneously. Male hippocampus exhibited a common effect of all three chemicals on genes involved in cell-autonomous processes, Sox6, Sox11, Pou2f2/Oct2, and Pou3f2/Brn2, all upregulated at the high dose. Additional genes of the Sox and Pou families were affected by only one or two of the chemicals. Real time RT PCR showed a comparable expression change for bisphenol A also at the lower dose. Female hippocampus exhibited much fewer genes with expression changes (almost none with false discovery rate <0.05), and none of the genes of the Sox and Pou families was affected. Since gene network analyses in male hippocampus suggested a link between Sox6 and miR-24, known to be repressed by activation of ER-alpha and to repress Sox6 in other tissues, this microRNA was measured. miR-24 was downregulated by all chemicals at the high dose in males. Values of Sox6 mRNA and miR-24 were inversely correlated in individual male hippocampus samples, supporting the hypothesis that the change in Sox6 expression resulted from an action of miR-24. In contrast, miR-24 levels remained unchanged in hippocampus of females. A sexually dimorphic response of miR-24 may thus be at the basis of the sex difference in Sox6 expression changes following exposure to the three chemicals. ER-alpha expression was also sex-dependent, but the expression changes did not parallel those of potential downstream genes such as Sox6. Sox6 is known to suppress differentiation of Parvalbumin (Pvalb)-expressing interneurons. Individual Sox6 levels (FPKM) were inversely correlated with levels of Pvalb, but not with markers of Sox6-independent interneuron subpopulations, Nos1 and 5HT3aR. Effects on interneuron development are further suggested, in males, by expression changes of Nrg1 and its receptor Erbb4, controlling interneuron migration. Our study disclosed new types of EDC-responsive morphogenetic genes, and illustrated the potential relevance of microRNAs in sexually dimorphic EDC actions.
Collapse
Affiliation(s)
- Walter Lichtensteiger
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Catherine Bassetti-Gaille
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center, Swiss Federal Institute of Technology and University of Zurich, Zurich, Switzerland
| | - Jelena Kühn Georgijevic
- Functional Genomics Center, Swiss Federal Institute of Technology and University of Zurich, Zurich, Switzerland
| | | | - Margret Schlumpf
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
Poleksic J, Aksic M, Kapor S, Aleksic D, Stojkovic T, Radovic M, Djulejic V, Markovic B, Stamatakis A. Effects of Maternal Deprivation on the Prefrontal Cortex of Male Rats: Cellular, Neurochemical, and Behavioral Outcomes. Front Behav Neurosci 2021; 15:666547. [PMID: 34819843 PMCID: PMC8606589 DOI: 10.3389/fnbeh.2021.666547] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 10/08/2021] [Indexed: 01/13/2023] Open
Abstract
Stressful events experienced during early life are associated with increased vulnerability of developing psychopathology in adulthood. In the present study, we exposed 9-day-old Wistar rats to 24 h maternal deprivation (MD) with the aim to investigate the impact of early life stress (ELS) on morphological, biochemical, and functional aspects of the prefrontal cortex (PFC), a brain region particularly sensitive to stress. We found that in the superficial medial orbital cortex (MO), young adult male rats had reduced density of GAD67 and CCK immunopositive cells, while the rostral part of the ventral lateral orbital cortex (roVLO) showed a decrease in the density of GAD67 immunopositive cells in both superficial and deep layers. In addition, the superficial rostral part of area 1 of the cingulate cortex (roCg1) and deep prelimbic cortex (PrL) was also affected by MD indicated by the reduction in PV immunopositive cellular density. Furthermore, MD induced upregulation of brain-derived neurotrophic factor (BDNF), while it did not affect the overall expression of Iba1 in neonatal or young adult PFC as measured by Western blot, however, microglial activation in young adult MD rats was detected immunohistochemically in deep layers of MO and infralimbic cortex (IL). Interestingly, when young adult male rats were subjected to a behavioral flexibility test in a T-maze, MD rats showed a subtle impairment in T-maze reversal learning indicating a mildly affected PFC function. Taken together, our findings demonstrated that MD reduced the density of interneurons and induced microglial activation, in particular, PFC areas at young adulthood, and could alter synaptic plasticity accompanied by PFC dysfunction.
Collapse
Affiliation(s)
- Joko Poleksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Aksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Slobodan Kapor
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dubravka Aleksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tihomir Stojkovic
- Institute of Clinical and Medical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marina Radovic
- Institute of Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Vuk Djulejic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Branka Markovic
- Faculty of Sport and Physical Education, University of Belgrade, Belgrade, Serbia
| | - Antonios Stamatakis
- Biology-Biochemistry Lab, School of Health Sciences, Faculty of Nursing, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
17
|
Prönneke A, Witte M, Möck M, Staiger JF. Neuromodulation Leads to a Burst-Tonic Switch in a Subset of VIP Neurons in Mouse Primary Somatosensory (Barrel) Cortex. Cereb Cortex 2021; 30:488-504. [PMID: 31210267 DOI: 10.1093/cercor/bhz102] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 12/14/2022] Open
Abstract
Neocortical GABAergic interneurons expressing vasoactive intestinal polypeptide (VIP) contribute to sensory processing, sensorimotor integration, and behavioral control. In contrast to other major subpopulations of GABAergic interneurons, VIP neurons show a remarkable diversity. Studying morphological and electrophysiological properties of VIP cells, we found a peculiar group of neurons in layer II/III of mouse primary somatosensory (barrel) cortex, which showed a highly dynamic burst firing behavior at resting membrane potential that switched to tonic mode at depolarized membrane potentials. Furthermore, we demonstrate that burst firing depends on T-type calcium channels. The burst-tonic switch could be induced by acetylcholine (ACh) and serotonin. ACh mediated a depolarization via nicotinic receptors whereas serotonin evoked a biphasic depolarization via ionotropic and metabotropic receptors in 48% of the population and a purely monophasic depolarization via metabotropic receptors in the remaining cells. These data disclose an electrophysiologically defined subpopulation of VIP neurons that via neuromodulator-induced changes in firing behavior is likely to regulate the state of cortical circuits in a profound manner.
Collapse
Affiliation(s)
- Alvar Prönneke
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität, D-37075 Göttingen, Germany
| | - Mirko Witte
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität, D-37075 Göttingen, Germany
| | - Martin Möck
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität, D-37075 Göttingen, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität, D-37075 Göttingen, Germany
| |
Collapse
|
18
|
Vasistha NA, Khodosevich K. The impact of (ab)normal maternal environment on cortical development. Prog Neurobiol 2021; 202:102054. [PMID: 33905709 DOI: 10.1016/j.pneurobio.2021.102054] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/01/2021] [Accepted: 04/20/2021] [Indexed: 12/24/2022]
Abstract
The cortex in the mammalian brain is the most complex brain region that integrates sensory information and coordinates motor and cognitive processes. To perform such functions, the cortex contains multiple subtypes of neurons that are generated during embryogenesis. Newly born neurons migrate to their proper location in the cortex, grow axons and dendrites, and form neuronal circuits. These developmental processes in the fetal brain are regulated to a large extent by a great variety of factors derived from the mother - starting from simple nutrients as building blocks and ending with hormones. Thus, when the normal maternal environment is disturbed due to maternal infection, stress, malnutrition, or toxic substances, it might have a profound impact on cortical development and the offspring can develop a variety of neurodevelopmental disorders. Here we first describe the major developmental processes which generate neuronal diversity in the cortex. We then review our knowledge of how most common maternal insults affect cortical development, perturb neuronal circuits, and lead to neurodevelopmental disorders. We further present a concept of selective vulnerability of cortical neuronal subtypes to maternal-derived insults, where the vulnerability of cortical neurons and their progenitors to an insult depends on the time (developmental period), place (location in the developing brain), and type (unique features of a cell type and an insult). Finally, we provide evidence for the existence of selective vulnerability during cortical development and identify the most vulnerable neuronal types, stages of differentiation, and developmental time for major maternal-derived insults.
Collapse
Affiliation(s)
- Navneet A Vasistha
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
19
|
Jun SB, Ikeda SR, Sung JE, Lovinger DM. Ethanol induces persistent potentiation of 5-HT 3 receptor-stimulated GABA release at synapses on rat hippocampal CA1 neurons. Neuropharmacology 2021; 184:108415. [PMID: 33275959 PMCID: PMC11009934 DOI: 10.1016/j.neuropharm.2020.108415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 11/18/2022]
Abstract
Several studies have shown that ethanol (EtOH) can enhance the activity of GABAergic synapses via presynaptic mechanisms, including in hippocampal CA1 neurons. The serotonin type 3 receptor (5-HT3-R) has been implicated in the neural actions of ethanol (EtOH) and in modulation of GABA release from presynaptic terminals. In the present study, we investigated EtOH modulation of GABA release induced by 5-HT3-R activation using the mechanically isolated neuron/bouton preparation from the rat CA1 hippocampal subregion. EtOH application before and during exposure to the selective 5-HT3 receptor agonist, m-chlorophenylbiguanide (mCPBG) potentiated the mCPBG-induced increases in the peak frequency and charge transfer of spontaneous GABAergic inhibitory postsynaptic currents. Interestingly, the potentiation was maintained even after EtOH was removed from the preparation. A protein kinase A inhibitor reduced the magnitude of EtOH potentiation. Fluorescent Ca2+ imaging showed that Ca2+ transients in the presynaptic terminals increased during EtOH exposure. These findings indicate that EtOH produces long-lasting potentiation of 5-HT3-induced GABA release by modulating calcium levels, via a process involving cAMP-mediated signaling in presynaptic terminals.
Collapse
Affiliation(s)
- Sang Beom Jun
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760, Republic of Korea; Smart Factory Multidisciplinary Program, Ewha Womans University, Seoul, 03760, Republic of Korea; Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
| | - Stephen R Ikeda
- Section on Transmitter Signaling, Laboratory of Molecular Physiology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Jee Eun Sung
- Department of Communication Disorders, Ewha Womans University, Seoul, 03760, Republic of Korea.
| | - David M Lovinger
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
20
|
PlexinA4-Semaphorin3A-mediated crosstalk between main cortical interneuron classes is required for superficial interneuron lamination. Cell Rep 2021; 34:108644. [PMID: 33503438 DOI: 10.1016/j.celrep.2020.108644] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/30/2020] [Accepted: 12/21/2020] [Indexed: 01/03/2023] Open
Abstract
In the mammalian cerebral cortex, the developmental events governing allocation of different classes of inhibitory interneurons (INs) to distinct cortical layers are poorly understood. Here we report that the guidance receptor PlexinA4 (PLXNA4) is upregulated in serotonin receptor 3a-expressing (HTR3A+) cortical INs (hINs) as they invade the cortical plate, and that it regulates their laminar allocation to superficial cortical layers. We find that the PLXNA4 ligand Semaphorin3A (SEMA3A) acts as a chemorepulsive factor on hINs migrating into the nascent cortex and demonstrate that SEMA3A specifically controls their laminar positioning through PLXNA4. We identify deep-layer INs as a major source of SEMA3A in the developing cortex and demonstrate that targeted genetic deletion of Sema3a in these INs specifically affects laminar allocation of hINs. These data show that, in the neocortex, deep-layer INs control laminar allocation of hINs into superficial layers.
Collapse
|
21
|
Shen W, Ba R, Su Y, Ni Y, Chen D, Xie W, Pleasure SJ, Zhao C. Foxg1 Regulates the Postnatal Development of Cortical Interneurons. Cereb Cortex 2020; 29:1547-1560. [PMID: 29912324 DOI: 10.1093/cercor/bhy051] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/23/2018] [Accepted: 02/15/2018] [Indexed: 12/12/2022] Open
Abstract
Abnormalities in cortical interneurons are closely associated with neurological diseases. Most patients with Foxg1 syndrome experience seizures, suggesting a possible role of Foxg1 in the cortical interneuron development. Here, by conditional deletion of Foxg1, which was achieved by crossing Foxg1fl/fl with the Gad2-CreER line, we found the postnatal distributions of somatostatin-, calretinin-, and neuropeptide Y-positive interneurons in the cortex were impaired. Further investigations revealed an enhanced dendritic complexity and decreased migration capacity of Foxg1-deficient interneurons, accompanied by remarkable downregulation of Dlx1 and CXCR4. Overexpression of Dlx1 or knock down its downstream Pak3 rescued the differentiation detects, demonstrated that Foxg1 functioned upstream of Dlx1-Pak3 signal pathway to regulate the postnatal development of cortical interneurons. Due to the imbalanced neural circuit, Foxg1 mutants showed increased seizure susceptibility. These findings will improve our understanding of the postnatal development of interneurons and help to elucidate the mechanisms underlying seizure in patients carrying Foxg1 mutations.
Collapse
Affiliation(s)
- Wei Shen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Ru Ba
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Yan Su
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Yang Ni
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Dongsheng Chen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Wei Xie
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Institute of Life Science, Southeast University, Nanjing, P. R. China
| | - Samuel J Pleasure
- Department of Neurology, Weill Institute for Neuroscience, Programs in Neuroscience and Developmental Stem Cell Biology, UCSF, San Francisco, CA, USA
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China.,Center of Depression, Beijing Institute for Brain Disorders, Beijing 100069, People's Republic of China
| |
Collapse
|
22
|
Asgarian Z, Magno L, Ktena N, Harris KD, Kessaris N. Hippocampal CA1 Somatostatin Interneurons Originate in the Embryonic MGE/POA. Stem Cell Reports 2019; 13:793-802. [PMID: 31631021 PMCID: PMC6895756 DOI: 10.1016/j.stemcr.2019.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 01/10/2023] Open
Abstract
Oriens lacunosum-moleculare (O-LM) interneurons constitute 40% of hippocampal interneurons expressing Somatostatin (SST). Recent evidence has indicated a dual origin for these cells in the medial and caudal ganglionic eminences (MGE and CGE), with expression of Htr3a as a distinguishing factor. This is strikingly different from cortical SST interneurons that have a single origin within the MGE/preoptic area (POA). We reassessed the origin of hippocampal SST interneurons using a range of genetic lineage-tracing mice combined with single-cell transcriptomic analysis. We find a common origin for all hippocampal SST interneurons in NKX2-1-expressing progenitors of the telencephalic neuroepithelium and an MGE/POA-like transcriptomic signature for all SST clusters. This suggests that functional heterogeneity within the SST CA1 population cannot be attributed to a differential MGE/CGE genetic origin.
Collapse
Affiliation(s)
- Zeinab Asgarian
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK; Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Lorenza Magno
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK; Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Niki Ktena
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Kenneth D Harris
- UCL Institute of Neurology at the Cruciform Building and Department of Neuroscience, Physiology, and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Nicoletta Kessaris
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK; Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
23
|
Winterer J, Lukacsovich D, Que L, Sartori AM, Luo W, Földy C. Single-cell RNA-Seq characterization of anatomically identified OLM interneurons in different transgenic mouse lines. Eur J Neurosci 2019; 50:3750-3771. [PMID: 31420995 PMCID: PMC6973274 DOI: 10.1111/ejn.14549] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 07/30/2019] [Accepted: 08/09/2019] [Indexed: 01/18/2023]
Abstract
Inhibitory GABAergic interneurons create different brain activity patterns that correlate with behavioural states. In this characterizing study, we used single-cell RNA-Seq to analyse anatomically- and electrophysiologically identified hippocampal oriens-lacunosum moleculare (OLM) interneurons. OLMs express somatostatin (Sst), generate feedback inhibition and play important roles in theta oscillations and fear encoding. Although an anatomically- and biophysically homogenous population, OLMs presumably comprise of two functionally distinct types with different developmental origins, inferred from the expression pattern of serotonin type-3a (5-HT3a, or Htr3a) receptor subunit and 5-HT excitability in a set of OLMs. To broadly characterize OLM cells, we used the Sst-Cre and the BAC transgenic Htr3a-Cre mouse lines and separately analysed SstCre-OLM and Htr3aCre-OLM types. We found a surprisingly consistent expression of Npy in OLMs, which was previously not associated with the identity of this type. Our analyses furthermore revealed uniform expression of developmental origin-related genes, including transcription factors and neurexin isoforms, without providing support for the current view that OLMs may originate from multiple neurogenic zones. Together, we found that OLMs constitute a highly homogenous transcriptomic population. Finally, our results revealed surprisingly infrequent expression of Htr3a in only ~10% of OLMs and an apparently specific expression of the 5-HT3b subunit-coding gene Htr3b in Htr3aCre-OLMs, but not in SstCre-OLMs. However, additional in situ hybridization experiments suggested that the differential expression of Htr3b may represent an unexpected consequence arising from the design of the Htr3a-Cre BAC transgenic line.
Collapse
Affiliation(s)
- Jochen Winterer
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Natural Sciences, University of Zürich, Zürich, Switzerland
| | - David Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Natural Sciences, University of Zürich, Zürich, Switzerland
| | - Lin Que
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Natural Sciences, University of Zürich, Zürich, Switzerland
| | - Andrea M Sartori
- Institute for Regenerative Medicine, Department of Health Sciences and Technology, ETH Zürich, University of Zürich, Zürich, Switzerland
| | - Wenshu Luo
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Natural Sciences, University of Zürich, Zürich, Switzerland
| | - Csaba Földy
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Natural Sciences, University of Zürich, Zürich, Switzerland
| |
Collapse
|
24
|
Duchatel RJ, Shannon Weickert C, Tooney PA. White matter neuron biology and neuropathology in schizophrenia. NPJ SCHIZOPHRENIA 2019; 5:10. [PMID: 31285426 PMCID: PMC6614474 DOI: 10.1038/s41537-019-0078-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/06/2019] [Indexed: 12/17/2022]
Abstract
Schizophrenia is considered a neurodevelopmental disorder as it often manifests before full brain maturation and is also a cerebral cortical disorder where deficits in GABAergic interneurons are prominent. Whilst most neurons are located in cortical and subcortical grey matter regions, a smaller population of neurons reside in white matter tracts of the primate and to a lesser extent, the rodent brain, subjacent to the cortex. These interstitial white matter neurons (IWMNs) have been identified with general markers for neurons [e.g., neuronal nuclear antigen (NeuN)] and with specific markers for neuronal subtypes such as GABAergic neurons. Studies of IWMNs in schizophrenia have primarily focused on their density underneath cortical areas known to be affected in schizophrenia such as the dorsolateral prefrontal cortex. Most of these studies of postmortem brains have identified increased NeuN+ and GABAergic IWMN density in people with schizophrenia compared to healthy controls. Whether IWMNs are involved in the pathogenesis of schizophrenia or if they are increased because of the cortical pathology in schizophrenia is unknown. We also do not understand how increased IWMN might contribute to brain dysfunction in the disorder. Here we review the literature on IWMN pathology in schizophrenia. We provide insight into the postulated functional significance of these neurons including how they may contribute to the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Ryan J Duchatel
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia
- Priority Centre for Brain and Mental Health Research and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, 2031, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, New York, 13210, USA
| | - Paul A Tooney
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Priority Centre for Brain and Mental Health Research and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
25
|
Emmenegger V, Qi G, Wang H, Feldmeyer D. Morphological and Functional Characterization of Non-fast-Spiking GABAergic Interneurons in Layer 4 Microcircuitry of Rat Barrel Cortex. Cereb Cortex 2019; 28:1439-1457. [PMID: 29329401 PMCID: PMC6093438 DOI: 10.1093/cercor/bhx352] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Indexed: 12/23/2022] Open
Abstract
GABAergic interneurons are notorious for their heterogeneity, despite constituting a small fraction of the neuronal population in the neocortex. Classification of interneurons is crucial for understanding their widespread cortical functions as they provide a complex and dynamic network, balancing excitation and inhibition. Here, we investigated different types of non-fast-spiking (nFS) interneurons in Layer 4 (L4) of rat barrel cortex using whole-cell patch-clamp recordings with biocytin-filling. Based on a quantitative analysis on a combination of morphological and electrophysiological parameters, we identified 5 distinct types of L4 nFS interneurons: 1) trans-columnar projecting interneurons, 2) locally projecting non-Martinotti-like interneurons, 3) supra-granular projecting Martinotti-like interneurons, 4) intra-columnar projecting VIP-like interneurons, and 5) locally projecting neurogliaform-like interneurons. Trans-columnar projecting interneurons are one of the most striking interneuron types, which have not been described so far in Layer 4. They feature extensive axonal collateralization not only in their home barrel but also in adjacent barrels. Furthermore, we identified that most of the L4 nFS interneurons express somatostatin, while few are positive for the transcription factor Prox1. The morphological and electrophysiological characterization of different L4 nFS interneuron types presented here provides insights into their synaptic connectivity and functional role in cortical information processing.
Collapse
Affiliation(s)
- Vishalini Emmenegger
- Institute of Neuroscience and Medicine, INM-2 and INM-10, Research Centre Jülich, Jülich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Department of Biosystems Sciences and Engineering, Bio Engineering Lab, ETH Zürich, Basel, Switzerland
| | - Guanxiao Qi
- Institute of Neuroscience and Medicine, INM-2 and INM-10, Research Centre Jülich, Jülich, Germany
| | - Haijun Wang
- Institute of Neuroscience and Medicine, INM-2 and INM-10, Research Centre Jülich, Jülich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- School of Electronic Engineering, Nanjing Xiaozhuang University, Nanjing, P.R. China
| | - Dirk Feldmeyer
- Institute of Neuroscience and Medicine, INM-2 and INM-10, Research Centre Jülich, Jülich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
- Jülich Aachen Research Alliance, Translational Brain Medicine (JARA Brain), Aachen, Germany
- Address correspondence to Dirk Feldmeyer, Institute of Neuroscience and Medicine (INM-2), Research Centre Jülich, D-52425 Jülich, Germany.
| |
Collapse
|
26
|
Wester JC, Mahadevan V, Rhodes CT, Calvigioni D, Venkatesh S, Maric D, Hunt S, Yuan X, Zhang Y, Petros TJ, McBain CJ. Neocortical Projection Neurons Instruct Inhibitory Interneuron Circuit Development in a Lineage-Dependent Manner. Neuron 2019; 102:960-975.e6. [PMID: 31027966 DOI: 10.1016/j.neuron.2019.03.036] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 02/13/2019] [Accepted: 03/25/2019] [Indexed: 12/30/2022]
Abstract
Neocortical circuits consist of stereotypical motifs that must self-assemble during development. Recent evidence suggests that the subtype identity of both excitatory projection neurons (PNs) and inhibitory interneurons (INs) is important for this process. We knocked out the transcription factor Satb2 in PNs to induce those of the intratelencephalic (IT) type to adopt a pyramidal tract (PT)-type identity. Loss of IT-type PNs selectively disrupted the lamination and circuit integration of INs derived from the caudal ganglionic eminence (CGE). Strikingly, reprogrammed PNs demonstrated reduced synaptic targeting of CGE-derived INs relative to controls. In control mice, IT-type PNs targeted neighboring CGE INs, while PT-type PNs did not in deep layers, confirming this lineage-dependent motif. Finally, single-cell RNA sequencing revealed that major CGE IN subtypes were conserved after loss of IT PNs, but with differential transcription of synaptic proteins and signaling molecules. Thus, IT-type PNs influence CGE-derived INs in a non-cell-autonomous manner during cortical development.
Collapse
Affiliation(s)
- Jason C Wester
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Vivek Mahadevan
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Christopher T Rhodes
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Daniela Calvigioni
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA; Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Sanan Venkatesh
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Steven Hunt
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Xiaoqing Yuan
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Yajun Zhang
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Timothy J Petros
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Chris J McBain
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Wang C, Yu B, Li M, Zhao C, Roper SN, Chen H. Two Groups of eGFP-Expressing Neurons with Distinct Characteristics in the Neocortex of GIN Mice. Neuroscience 2019; 404:268-281. [PMID: 30703506 DOI: 10.1016/j.neuroscience.2019.01.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 12/20/2022]
Abstract
GIN (GFP-expressing inhibitory interneuron) transgenic mice are believed to express the enhanced GFP (eGFP) in a subset of somatostatin (SST)-expressing interneurons in the neocortex and have been widely used in the study on SST interneurons. Previous studies showed that eGFP+ neurons in the neocortex are distributed in the layer II-IV and upper layer V (cortical eGFP neurons) and contain SST. In this study, we reported a new group of eGFP+ neurons in GIN mice at early postnatal ages, which was located in the deep layer of the lateral neocortex as clusters (cluster eGFP neurons). Cluster eGFP neurons were noticeable at birth but disappeared within two months, in contrast to cortical eGFP neurons that started to appear around postnatal day 3 to 5 and existed through life. Cluster eGFP neurons were not immunoreactive for SST antibodies, contrary to cortical eGFP neurons. They were also not immunolabeled by parvalbumin, a marker for another major type of interneurons, and Ca2+/calmodulin-dependent kinases II, a commonly used marker for excitatory neurons. Firing rate, afterhyperpolarization, and excitatory synaptic activity significantly enhanced in cortical eGFP neurons during postnatal development, but these properties remained mostly unchanged in cluster eGFP neurons. Short-term plasticity of the excitatory synapse showed robust facilitation in cortical eGFP neurons but depression in cluster eGFP neurons. These results implied that eGFP might also be expressed in other types of cortical neurons in addition to SST-containing interneurons in GIN mice at early postnatal ages.
Collapse
Affiliation(s)
- Chunlian Wang
- Key Lab of Cognition and Personality of the Ministry of Education, Collaborative Innovation Center for Brain Science, School of Psychology, Southwest University, Chongqing, China
| | - Baocong Yu
- Key Lab of Developmental Genes and Human Diseases of the Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Meiyi Li
- Key Lab of Cognition and Personality of the Ministry of Education, Collaborative Innovation Center for Brain Science, School of Psychology, Southwest University, Chongqing, China
| | - Chunjie Zhao
- Key Lab of Developmental Genes and Human Diseases of the Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Steven N Roper
- Department of Neurosurgery, University of Florida, Gainesville, USA
| | - Huanxin Chen
- Key Lab of Cognition and Personality of the Ministry of Education, Collaborative Innovation Center for Brain Science, School of Psychology, Southwest University, Chongqing, China.
| |
Collapse
|
28
|
Keller D, Erö C, Markram H. Cell Densities in the Mouse Brain: A Systematic Review. Front Neuroanat 2018; 12:83. [PMID: 30405363 PMCID: PMC6205984 DOI: 10.3389/fnana.2018.00083] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/20/2018] [Indexed: 11/29/2022] Open
Abstract
The mouse brain is the most extensively studied brain of all species. We performed an exhaustive review of the literature to establish our current state of knowledge on cell numbers in mouse brain regions, arguably the most fundamental property to measure when attempting to understand a brain. The synthesized information, collected in one place, can be used by both theorists and experimentalists. Although for commonly-studied regions cell densities could be obtained for principal cell types, overall we know very little about how many cells are present in most brain regions and even less about cell-type specific densities. There is also substantial variation in cell density values obtained from different sources. This suggests that we need a new approach to obtain cell density datasets for the mouse brain.
Collapse
Affiliation(s)
- Daniel Keller
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | | | | |
Collapse
|
29
|
Bertacchi M, Parisot J, Studer M. The pleiotropic transcriptional regulator COUP-TFI plays multiple roles in neural development and disease. Brain Res 2018; 1705:75-94. [PMID: 29709504 DOI: 10.1016/j.brainres.2018.04.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 12/23/2022]
Abstract
Transcription factors are expressed in a dynamic fashion both in time and space during brain development, and exert their roles by activating a cascade of multiple target genes. This implies that understanding the precise function of a transcription factor becomes a challenging task. In this review, we will focus on COUP-TFI (or NR2F1), a nuclear receptor belonging to the superfamily of the steroid/thyroid hormone receptors, and considered to be one of the major transcriptional regulators orchestrating cortical arealization, cell-type specification and maturation. Recent data have unraveled the multi-faceted functions of COUP-TFI in the development of several mouse brain structures, including the neocortex, hippocampus and ganglionic eminences. Despite NR2F1 mutations and deletions in humans have been linked to a complex neurodevelopmental disease mainly associated to optic atrophy and intellectual disability, its role during the formation of the retina and optic nerve remains unclear. In light of its major influence in cortical development, we predict that its haploinsufficiency might be the cause of other cognitive diseases, not identified so far. Mouse models offer a unique opportunity of dissecting COUP-TFI function in different regions during brain assembly; hence, the importance of comparing and discussing common points linking mouse models to human patients' symptoms.
Collapse
Affiliation(s)
- Michele Bertacchi
- Université Côte d'Azur, CNRS, Inserm, iBV - Institut de Biologie Valrose, 06108 Nice, France.
| | - Josephine Parisot
- Université Côte d'Azur, CNRS, Inserm, iBV - Institut de Biologie Valrose, 06108 Nice, France
| | - Michèle Studer
- Université Côte d'Azur, CNRS, Inserm, iBV - Institut de Biologie Valrose, 06108 Nice, France.
| |
Collapse
|
30
|
Kondo M, Koyama Y, Nakamura Y, Shimada S. A novel 5HT3 receptor-IGF1 mechanism distinct from SSRI-induced antidepressant effects. Mol Psychiatry 2018; 23:833-842. [PMID: 28439104 DOI: 10.1038/mp.2017.87] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 03/16/2017] [Accepted: 03/21/2017] [Indexed: 01/14/2023]
Abstract
Depression is a common mental disorder affecting around 350 million people worldwide. Although selective serotonin reuptake inhibitors (SSRIs) are the most widely used antidepressants, a significant proportion of depressed patients do not achieve remission with SSRIs. In this study, we show that a serotonin type 3 receptor (5HT3R) agonist induces antidepressant effects as well as hippocampal neurogenesis independent of fluoxetine (a commonly used SSRI). Notably, our histological analysis reveals that 5HT3R and insulin-like growth factor 1 (IGF1) are expressed in the same neurons in the subgranular zone of the hippocampal dentate gyrus. Furthermore, our in vivo microdialysis analysis shows that 5HT3R regulates hippocampal extracellular IGF1 levels, and we also show that 5HT3R-dependent hippocampal neurogenesis is mediated by increased IGF1 levels. Altogether, our findings suggest a novel 5HT3R-IGF1 mechanism that is distinct from fluoxetine-induced responses and that provides a new therapeutic target for depression, especially bringing significant benefits for SSRI-resistant depressed patients.
Collapse
Affiliation(s)
- M Kondo
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Y Koyama
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Y Nakamura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - S Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
31
|
Niquille M, Limoni G, Markopoulos F, Cadilhac C, Prados J, Holtmaat A, Dayer A. Neurogliaform cortical interneurons derive from cells in the preoptic area. eLife 2018; 7:32017. [PMID: 29557780 PMCID: PMC5860868 DOI: 10.7554/elife.32017] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/21/2018] [Indexed: 12/18/2022] Open
Abstract
Delineating the basic cellular components of cortical inhibitory circuits remains a fundamental issue in order to understand their specific contributions to microcircuit function. It is still unclear how current classifications of cortical interneuron subtypes relate to biological processes such as their developmental specification. Here we identified the developmental trajectory of neurogliaform cells (NGCs), the main effectors of a powerful inhibitory motif recruited by long-range connections. Using in vivo genetic lineage-tracing in mice, we report that NGCs originate from a specific pool of 5-HT3AR-expressing Hmx3+ cells located in the preoptic area (POA). Hmx3-derived 5-HT3AR+ cortical interneurons (INs) expressed the transcription factors PROX1, NR2F2, the marker reelin but not VIP and exhibited the molecular, morphological and electrophysiological profile of NGCs. Overall, these results indicate that NGCs are a distinct class of INs with a unique developmental trajectory and open the possibility to study their specific functional contribution to cortical inhibitory microcircuit motifs. Our brain contains over a 100 billion nerve cells or neurons, and each of them is thought to connect to over 1,000 other neurons. Together, these cells form a complex network to convey information from our surroundings or transmit messages to designated destinations. This circuitry forms the basis of our unique cognitive abilities. In the cerebral cortex – the largest region of the brain – two main types of neurons can be found: projection neurons, which transfer information to other regions in the brain, and interneurons, which connect locally to different neurons and harmonize this information by inhibiting specific messages. The over 20 different types of known interneurons come in different shapes and properties and are thought to play a key role in powerful computations such as learning and memory. Since interneurons are hard to track, it is still unclear when and how they start to form and mature as the brain of an embryo develops. For example, one type of interneurons called the neurogliaform cells, have a very distinct shape and properties. But, until now, the origin of this cell type had been unknown. To find out how neurogliaform cells develop, Niquille, Limoni, Markopoulos et al. used a specific gene called Hmx3 to track these cells over time. With this strategy, the shapes and properties of the cells could be analyzed. The results showed that neurogliaform cells originate from a region outside of the cerebral cortex called the preoptic area, and later travel over long distances to reach their final location. The cells reach the cortex a few days after their birth and take several weeks to mature. These results suggest that the traits of a specific type of neuron is determined very early in life. By labeling this unique subset of interneurons, researchers will now be able to identify the specific molecular mechanisms that help the neurogliaform cells to develop. Furthermore, it will provide a new strategy to fully understand what role these cells play in processing information and guiding behavior.
Collapse
Affiliation(s)
- Mathieu Niquille
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | - Greta Limoni
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | - Foivos Markopoulos
- Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | - Christelle Cadilhac
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | - Julien Prados
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | - Anthony Holtmaat
- Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | - Alexandre Dayer
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| |
Collapse
|
32
|
Bohannon AS, Hablitz JJ. Optogenetic dissection of roles of specific cortical interneuron subtypes in GABAergic network synchronization. J Physiol 2018; 596:901-919. [PMID: 29274075 PMCID: PMC5830415 DOI: 10.1113/jp275317] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/13/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS An increase in the excitability of GABAergic cells has typically been assumed to decrease network activity, potentially producing overall anti-epileptic effects. Recent data suggest that inhibitory networks may actually play a role in initiating epileptiform activity. We show that activation of GABAergic interneurons can elicit synchronous long-lasting network activity. Specific interneuron subpopulations differentially contributed to GABA network synchrony, indicating cell type-specific contributions of interneurons to cortical network activity. Interneurons may critically contribute to the generation of aberrant network activity characteristic of epilepsy, warranting further investigation into the contribution of distinct cortical interneuron subpopulations to the propagation and rhythmicity of epileptiform activity. ABSTRACT In the presence of the A-type K+ channel blocker 4-aminopyrdine, spontaneous synchronous network activity develops in the neocortex of mice of either sex. This aberrant synchrony persists in the presence of excitatory amino acid receptor antagonists (EAA blockers) and is considered to arise from synchronous firing of cortical interneurons (INs). Although much attention has been given to the mechanisms underlying this GABAergic synchrony, the contribution of specific IN subtypes to the generation of these long-lasting discharges (LLDs) is incompletely understood. We employed genetically-encoded channelrhodopsin and archaerhodopsin opsins to investigate the sufficiency and necessity, respectively, of activation of parvalbumin (PV), somatostatin (SST) and vasointestinal peptide (VIP)-expressing INs for the generation of synchronous neocortical GABAergic discharges. We found light-induced activation of PV or SST INs to be equally sufficient for the generation of LLDs, whereas activation of VIP INs was not. By contrast, light-induced inhibition of PV INs strongly reduced LLD initiation, whereas suppression of SST or VIP IN activity only partially attenuated LLD magnitude. These results suggest neocortical INs perform cell type-specific roles in the generation of aberrant GABAergic cortical network activity.
Collapse
Affiliation(s)
- Andrew S. Bohannon
- Department of NeurobiologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - John J. Hablitz
- Department of NeurobiologyUniversity of Alabama at BirminghamBirminghamALUSA
| |
Collapse
|
33
|
Ruiz-Reig N, Studer M. Rostro-Caudal and Caudo-Rostral Migrations in the Telencephalon: Going Forward or Backward? Front Neurosci 2017; 11:692. [PMID: 29311773 PMCID: PMC5742585 DOI: 10.3389/fnins.2017.00692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 11/13/2022] Open
Abstract
The generation and differentiation of an appropriate number of neurons, as well as its distribution in different parts of the brain, is crucial for the proper establishment, maintenance and plasticity of neural circuitries. Newborn neurons travel along the brain in a process known as neuronal migration, to finalize their correct position in the nervous system. Defects in neuronal migration produce abnormalities in the brain that can generate neurodevelopmental pathologies, such as autism, schizophrenia and intellectual disability. In this review, we present an overview of the developmental origin of the different telencephalic subdivisions and a description of migratory pathways taken by distinct neural populations traveling long distances before reaching their target position in the brain. In addition, we discuss some of the molecules implicated in the guidance of these migratory paths and transcription factors that contribute to the correct migration and integration of these neurons.
Collapse
|
34
|
Traub RD, Whittington MA, Hall SP. Does Epileptiform Activity Represent a Failure of Neuromodulation to Control Central Pattern Generator-Like Neocortical Behavior? Front Neural Circuits 2017; 11:78. [PMID: 29093667 PMCID: PMC5651241 DOI: 10.3389/fncir.2017.00078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/04/2017] [Indexed: 12/22/2022] Open
Abstract
Rhythmic motor patterns in invertebrates are often driven by specialized “central pattern generators” (CPGs), containing small numbers of neurons, which are likely to be “identifiable” in one individual compared with another. The dynamics of any particular CPG lies under the control of modulatory substances, amines, or peptides, entering the CPG from outside it, or released by internal constituent neurons; consequently, a particular CPG can generate a given rhythm at different frequencies and amplitudes, and perhaps even generate a repertoire of distinctive patterns. The mechanisms exploited by neuromodulators in this respect are manifold: Intrinsic conductances (e.g., calcium, potassium channels), conductance state of postsynaptic receptors, degree of plasticity, and magnitude and kinetics of transmitter release can all be affected. The CPG concept has been generalized to vertebrate motor pattern generating circuits (e.g., for locomotion), which may contain large numbers of neurons – a construct that is sensible, if there is enough redundancy: that is, the large number of neurons consists of only a small number of classes, and the cells within any one class act stereotypically. Here we suggest that CPG and modulator ideas may also help to understand cortical oscillations, normal ones, and particularly transition to epileptiform pathology. Furthermore, in the case illustrated, the mechanism of the transition appears to be an exaggerated form of a normal modulatory action used to influence sensory processing.
Collapse
Affiliation(s)
- Roger D Traub
- Department of Physical Sciences, IBM Thomas J. Watson Research Center, New York City, NY, United States
| | - Miles A Whittington
- Department of Biology, Hull York Medical School, University of York, York, United Kingdom
| | - Stephen P Hall
- Department of Biology, Hull York Medical School, University of York, York, United Kingdom
| |
Collapse
|
35
|
Pelkey KA, Chittajallu R, Craig MT, Tricoire L, Wester JC, McBain CJ. Hippocampal GABAergic Inhibitory Interneurons. Physiol Rev 2017; 97:1619-1747. [PMID: 28954853 DOI: 10.1152/physrev.00007.2017] [Citation(s) in RCA: 495] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/16/2017] [Accepted: 05/26/2017] [Indexed: 12/11/2022] Open
Abstract
In the hippocampus GABAergic local circuit inhibitory interneurons represent only ~10-15% of the total neuronal population; however, their remarkable anatomical and physiological diversity allows them to regulate virtually all aspects of cellular and circuit function. Here we provide an overview of the current state of the field of interneuron research, focusing largely on the hippocampus. We discuss recent advances related to the various cell types, including their development and maturation, expression of subtype-specific voltage- and ligand-gated channels, and their roles in network oscillations. We also discuss recent technological advances and approaches that have permitted high-resolution, subtype-specific examination of their roles in numerous neural circuit disorders and the emerging therapeutic strategies to ameliorate such pathophysiological conditions. The ultimate goal of this review is not only to provide a touchstone for the current state of the field, but to help pave the way for future research by highlighting where gaps in our knowledge exist and how a complete appreciation of their roles will aid in future therapeutic strategies.
Collapse
Affiliation(s)
- Kenneth A Pelkey
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ramesh Chittajallu
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Michael T Craig
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ludovic Tricoire
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Jason C Wester
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Chris J McBain
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| |
Collapse
|
36
|
Turrero García M, Harwell CC. Radial glia in the ventral telencephalon. FEBS Lett 2017; 591:3942-3959. [PMID: 28862741 DOI: 10.1002/1873-3468.12829] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 12/31/2022]
Abstract
The ventral telencephalon is the developmental origin of the basal ganglia and the source of neuronal and glial cells that integrate into developing circuits in other areas of the brain. Radial glia in the embryonic subpallium give rise to an enormous diversity of mature cell types, either directly or through other transit-amplifying progenitors. Here, we review current knowledge about these subpallial neural stem cells and their progeny, focusing on the period of neurogenesis. We describe their cell biological features and the extrinsic and intrinsic molecular codes that guide their fate specification in defined temporal and spatial sequences. We also discuss the role of clonal lineage in the organization and specification of mature neurons.
Collapse
Affiliation(s)
| | - Corey C Harwell
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
Regional Cellular Environment Shapes Phenotypic Variations of Hippocampal and Neocortical Chandelier Cells. J Neurosci 2017; 37:9901-9916. [PMID: 28912162 DOI: 10.1523/jneurosci.0047-17.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 08/26/2017] [Accepted: 09/01/2017] [Indexed: 11/21/2022] Open
Abstract
Different cortical regions processing distinct information, such as the hippocampus and the neocortex, share common cellular components and circuit motifs but form unique networks by modifying these cardinal units. Cortical circuits include diverse types of GABAergic interneurons (INs) that shape activity of excitatory principal neurons (PNs). Canonical IN types conserved across distinct cortical regions have been defined by their morphological, electrophysiological, and neurochemical properties. However, it remains largely unknown whether canonical IN types undergo specific modifications in distinct cortical regions and display "regional variants." It is also poorly understood whether such phenotypic variations are shaped by early specification or regional cellular environment. The chandelier cell (ChC) is a highly stereotyped IN type that innervates axon initial segments of PNs and thus serves as a good model with which to address this issue. Here, we show that Cadherin-6 (Cdh6), a homophilic cell adhesion molecule, is a reliable marker of ChCs and Cdh6-CreER mice (both sexes) provide genetic access to hippocampal ChCs (h-ChCs). We demonstrate that, compared with neocortical ChCs (nc-ChCs), h-ChCs cover twice as much area and innervate twice as many PNs. Interestingly, a subclass of h-ChCs exhibits calretinin (CR) expression, which is not found in nc-ChCs. Furthermore, we find that h-ChCs appear to be born earlier than nc-ChCs. Surprisingly, despite the difference in temporal origins, ChCs display host-region-dependent axonal/synaptic organization and CR expression when transplanted heterotopically. These results suggest that local cellular environment plays a critical role in shaping terminal phenotypes of regional IN variants in the hippocampus and the neocortex.SIGNIFICANCE STATEMENT Canonical interneuron (IN) types conserved across distinct cortical regions such as the hippocampus and the neocortex are defined by morphology, physiology, and gene expression. However, it remains unknown whether they display phenotypic variations in different cortical regions. In addition, it is unclear whether terminal phenotypes of regional IN variants belonging to a canonical IN type are determined intrinsically or extrinsically. Our results provide evidence of striking differences in axonal/synaptic organization and calretinin expression between hippocampal chandelier cells (ChCs) and neocortical ChCs. They also reveal that local cellular environment in distinct cortical regions regulates these terminal phenotypes. Therefore, our study suggests that local cortical environment shapes the phenotypes of regional IN variants, which may be required for unique circuit operations in distinct cortical regions.
Collapse
|
38
|
Afferent specific role of NMDA receptors for the circuit integration of hippocampal neurogliaform cells. Nat Commun 2017; 8:152. [PMID: 28751664 PMCID: PMC5532276 DOI: 10.1038/s41467-017-00218-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 06/13/2017] [Indexed: 01/28/2023] Open
Abstract
Appropriate integration of GABAergic interneurons into nascent cortical circuits is critical for ensuring normal information processing within the brain. Network and cognitive deficits associated with neurological disorders, such as schizophrenia, that result from NMDA receptor-hypofunction have been mainly attributed to dysfunction of parvalbumin-expressing interneurons that paradoxically express low levels of synaptic NMDA receptors. Here, we reveal that throughout postnatal development, thalamic, and entorhinal cortical inputs onto hippocampal neurogliaform cells are characterized by a large NMDA receptor-mediated component. This NMDA receptor-signaling is prerequisite for developmental programs ultimately responsible for the appropriate long-range AMPAR-mediated recruitment of neurogliaform cells. In contrast, AMPAR-mediated input at local Schaffer-collateral synapses on neurogliaform cells remains normal following NMDA receptor-ablation. These afferent specific deficits potentially impact neurogliaform cell mediated inhibition within the hippocampus and our findings reveal circuit loci implicating this relatively understudied interneuron subtype in the etiology of neurodevelopmental disorders characterized by NMDA receptor-hypofunction.Proper brain function depends on the correct assembly of excitatory and inhibitory neurons into neural circuits. Here the authors show that during early postnatal development in mice, NMDAR signaling via activity of long-range synaptic inputs onto neurogliaform cells is required for their appropriate integration into the hippocampal circuitry.
Collapse
|
39
|
Bellono NW, Bayrer JR, Leitch DB, Castro J, Zhang C, O'Donnell TA, Brierley SM, Ingraham HA, Julius D. Enterochromaffin Cells Are Gut Chemosensors that Couple to Sensory Neural Pathways. Cell 2017; 170. [PMID: 28648659 PMCID: PMC5839326 DOI: 10.1016/j.cell.2017.05.034] [Citation(s) in RCA: 532] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dietary, microbial, and inflammatory factors modulate the gut-brain axis and influence physiological processes ranging from metabolism to cognition. The gut epithelium is a principal site for detecting such agents, but precisely how it communicates with neural elements is poorly understood. Serotonergic enterochromaffin (EC) cells are proposed to fulfill this role by acting as chemosensors, but understanding how these rare and unique cell types transduce chemosensory information to the nervous system has been hampered by their paucity and inaccessibility to single-cell measurements. Here, we circumvent this limitation by exploiting cultured intestinal organoids together with single-cell measurements to elucidate intrinsic biophysical, pharmacological, and genetic properties of EC cells. We show that EC cells express specific chemosensory receptors, are electrically excitable, and modulate serotonin-sensitive primary afferent nerve fibers via synaptic connections, enabling them to detect and transduce environmental, metabolic, and homeostatic information from the gut directly to the nervous system.
Collapse
Affiliation(s)
- Nicholas W Bellono
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - James R Bayrer
- Department of Pediatrics, Division of Gastroenterology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Duncan B Leitch
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joel Castro
- Visceral Pain Group, Flinders University, Bedford Park, SA 5042, Australia; Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA 5000, Australia
| | - Chuchu Zhang
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tracey A O'Donnell
- Visceral Pain Group, Flinders University, Bedford Park, SA 5042, Australia; Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA 5000, Australia
| | - Stuart M Brierley
- Visceral Pain Group, Flinders University, Bedford Park, SA 5042, Australia; Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA 5000, Australia
| | - Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - David Julius
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
40
|
Transplantation of GABAergic interneurons for cell-based therapy. PROGRESS IN BRAIN RESEARCH 2017; 231:57-85. [PMID: 28554401 DOI: 10.1016/bs.pbr.2016.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Many neurological disorders stem from defects in or the loss of specific neurons. Neuron transplantation has tremendous clinical potential for central nervous system therapy as it may allow for the targeted replacement of those cells that are lost in diseases. Normally, most neurons are added during restricted periods of embryonic and fetal development. The permissive milieu of the developing brain promotes neuronal migration, neuronal differentiation, and synaptogenesis. Once this active period of neurogenesis ends, the chemical and physical environment of the brain changes dramatically. The brain parenchyma becomes highly packed with neuronal and glial processes, extracellular matrix, myelin, and synapses. The migration of grafted cells to allow them to home into target regions and become functionally integrated is a key challenge to neuronal transplantation. Interestingly, transplanted young telencephalic inhibitory interneurons are able to migrate, differentiate, and integrate widely throughout the postnatal brain. These grafted interneurons can also functionally modify local circuit activity. These features have facilitated the use of interneuron transplantation to study fundamental neurodevelopmental processes including cell migration, cell specification, and programmed neuronal cell death. Additionally, these cells provide a unique opportunity to develop interneuron-based strategies for the treatment of diseases linked to interneuron dysfunction and neurological disorders associated to circuit hyperexcitability.
Collapse
|
41
|
Koyama Y, Kondo M, Shimada S. Building a 5-HT3A Receptor Expression Map in the Mouse Brain. Sci Rep 2017; 7:42884. [PMID: 28276429 PMCID: PMC5343592 DOI: 10.1038/srep42884] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/16/2017] [Indexed: 01/07/2023] Open
Abstract
Of the many serotonin receptors, the type 3 receptors (5-HT3R) are the only ionotropic ones, playing a key role in fast synaptic transmission and cognitive and emotional brain function through controlled neuronal excitation. To better understand the various functions of 5-HT3Rs, it is very important to know their expression pattern in the central nervous system (CNS). To date, many distributional studies have shown localized 5-HT3R expression in the brain and spinal cord. However, an accurate pattern of 5-HT3R expression in the CNS remains to be elucidated. To investigate the distribution of 5-HT3R in the mouse brain in detail, we performed immunofluorescent staining using 5-HT3AR-GFP transgenic mice. We found strong 5-HT3AR expression in the olfactory bulb, cerebral cortex, hippocampus, and amygdala; and partial expression in the pons, medulla, and spinal cord. Meanwhile, the thalamus, hypothalamus, and midbrain exhibited a few 5-HT3AR-expressing cells, and no expression was detected in the cerebellum. Further, double-immunostaining using neural markers confirmed that 5-HT3AR is expressed in GABAergic interneurons containing somatostatin or calretinin. In the present study, we built a 5-HT3AR expression map in the mouse brain. Our findings make significant contributions in elucidating the novel functions of 5-HT3R in the CNS.
Collapse
Affiliation(s)
- Yoshihisa Koyama
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Makoto Kondo
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| |
Collapse
|
42
|
Finger TE, Bartel DL, Shultz N, Goodson NB, Greer CA. 5HTR3A-driven GFP labels immature olfactory sensory neurons. J Comp Neurol 2017; 525:1743-1755. [PMID: 28152579 DOI: 10.1002/cne.24180] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 01/25/2017] [Accepted: 01/26/2017] [Indexed: 12/12/2022]
Abstract
The ionotropic serotonin receptor, 5-HT3 , is expressed by many developing neurons within the central nervous system. Since the olfactory epithelium continues to generate new olfactory sensory neurons (OSNs) throughout life, we investigated the possibility that 5-HT3 is expressed in the adult epithelium. Using a transgenic mouse in which the promoter for the 5-HT3a subunit drives expression of green fluorescent protein (GFP), we assessed the expression of this marker in the olfactory epithelium of adult mice. Both the native 5-HT3a mRNA and GFP are expressed within globose basal cells of the olfactory and vomeronasal epithelium in adult mice. Whereas the 5-HT3a mRNA disappears relatively quickly after final cell division, the GFP label persists for about 5 days, thereby labeling immature OSNs in both the main olfactory system and vomeronasal organ. The GFP-labeled cells include both proliferative globose basal cells as well as immature OSNs exhibiting the hallmarks of ongoing differentiation including GAP43, PGP9.5, but the absence of olfactory marker protein. Some of the GFP-labeled OSNs show characteristics of more mature yet still developing OSNs including the presence of cilia extending from the apical knob and expression of NaV1.5, a component of the transduction cascade. These findings suggest that 5-HT3a is indicative of a proliferative or developmental state, regardless of age, and that the 5-HT3A GFP mice may prove useful for future studies of neurogenesis in the olfactory epithelium. J. Comp. Neurol. 525:1743-1755, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Thomas E Finger
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, Colorado.,Program in Neuroscience, University of Colorado School of Medicine, Aurora, Colorado
| | - Dianna L Bartel
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, Colorado.,Program in Neuroscience, University of Colorado School of Medicine, Aurora, Colorado.,Departments of Neurosurgery and Neuroscience, Yale University School of Medicine, New Haven, Connecticut
| | - Nicole Shultz
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, Colorado
| | - Noah B Goodson
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, Colorado.,Program in Neuroscience, University of Colorado School of Medicine, Aurora, Colorado
| | - Charles A Greer
- Departments of Neurosurgery and Neuroscience, Yale University School of Medicine, New Haven, Connecticut.,Interdepartmental Neuroscience Graduate Program, Yale University, New Haven, Connecticut
| |
Collapse
|
43
|
Frazer S, Prados J, Niquille M, Cadilhac C, Markopoulos F, Gomez L, Tomasello U, Telley L, Holtmaat A, Jabaudon D, Dayer A. Transcriptomic and anatomic parcellation of 5-HT 3AR expressing cortical interneuron subtypes revealed by single-cell RNA sequencing. Nat Commun 2017; 8:14219. [PMID: 28134272 PMCID: PMC5290279 DOI: 10.1038/ncomms14219] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 12/08/2016] [Indexed: 11/09/2022] Open
Abstract
Cortical GABAergic interneurons constitute a highly diverse population of inhibitory neurons that are key regulators of cortical microcircuit function. An important and heterogeneous group of cortical interneurons specifically expresses the serotonin receptor 3A (5-HT3AR) but how this diversity emerges during development is poorly understood. Here we use single-cell transcriptomics to identify gene expression patterns operating in Htr3a-GFP+ interneurons during early steps of cortical circuit assembly. We identify three main molecular types of Htr3a-GFP+ interneurons, each displaying distinct developmental dynamics of gene expression. The transcription factor Meis2 is specifically enriched in a type of Htr3a-GFP+ interneurons largely confined to the cortical white matter. These MEIS2-expressing interneurons appear to originate from a restricted region located at the embryonic pallial–subpallial boundary. Overall, this study identifies MEIS2 as a subclass-specific marker for 5-HT3AR-containing interstitial interneurons and demonstrates that the transcriptional and anatomical parcellation of cortical interneurons is developmentally coupled. Cortical GABAergic interneurons are highly diverse in their gene expression, electrophysiological properties, and connectivity. Here the authors reveal three distinct subtypes of Htr3a-GFP+ interneurons using the single-cell RNA-seq approach, and identify MEIS2 as a marker for one such subtype.
Collapse
Affiliation(s)
- Sarah Frazer
- Department of Psychiatry, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| | - Julien Prados
- Department of Psychiatry, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| | - Mathieu Niquille
- Department of Psychiatry, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| | - Christelle Cadilhac
- Department of Psychiatry, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| | - Foivos Markopoulos
- Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| | - Lucia Gomez
- Department of Psychiatry, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| | - Ugo Tomasello
- Department of Psychiatry, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| | - Ludovic Telley
- Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| | - Anthony Holtmaat
- Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| | - Alexandre Dayer
- Department of Psychiatry, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland.,Department of Basic Neurosciences, University of Geneva Medical School, Geneva 4 CH-1211, Switzerland
| |
Collapse
|
44
|
Ritter KE, Southard-Smith EM. Dynamic Expression of Serotonin Receptor 5-HT3A in Developing Sensory Innervation of the Lower Urinary Tract. Front Neurosci 2017; 10:592. [PMID: 28111539 PMCID: PMC5216032 DOI: 10.3389/fnins.2016.00592] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022] Open
Abstract
Sensory afferent signaling is required for normal function of the lower urinary tract (LUT). Despite the wide prevalence of bladder dysfunction and pelvic pain syndromes, few effective treatment options are available. Serotonin receptor 5-HT3A is a known mediator of visceral afferent signaling and has been implicated in bladder function. However, basic expression patterns for this gene and others among developing bladder sensory afferents that could be used to inform regenerative efforts aimed at treating deficiencies in pelvic innervation are lacking. To gain greater insight into the molecular characteristics of bladder sensory innervation, we conducted a thorough characterization of Htr3a expression in developing and adult bladder-projecting lumbosacral dorsal root ganglia (DRG) neurons. Using a transgenic Htr3a-EGFP reporter mouse line, we identified 5-HT3A expression at 10 days post coitus (dpc) in neural crest derivatives and in 12 dpc lumbosacral DRG. Using immunohistochemical co-localization we observed Htr3a-EGFP expression in developing lumbosacral DRG that partially coincides with neuropeptides CGRP and Substance P and capsaicin receptor TRPV1. A majority of Htr3a-EGFP+ DRG neurons also express a marker of myelinated Aδ neurons, NF200. There was no co-localization of 5-HT3A with the TRPV4 receptor. We employed retrograde tracing in adult Htr3a-EGFP mice to quantify the contribution of 5-HT3A+ DRG neurons to bladder afferent innervation. We found that 5-HT3A is expressed in a substantial proportion of retrograde traced DRG neurons in both rostral (L1, L2) and caudal (L6, S1) axial levels that supply bladder innervation. Most bladder-projecting Htr3a-EGFP+ neurons that co-express CGRP, Substance P, or TRPV1 are found in L1, L2 DRG, whereas Htr3a-EGFP+, NF200+ bladder-projecting neurons are from the L6, S1 axial levels. Our findings contribute much needed information regarding the development of LUT innervation and highlight the 5-HT3A serotonin receptor as a candidate for future studies of neurally mediated bladder control.
Collapse
Affiliation(s)
- K Elaine Ritter
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine Nashville, TN, USA
| | - E Michelle Southard-Smith
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine Nashville, TN, USA
| |
Collapse
|
45
|
Ansen-Wilson LJ, Lipinski RJ. Gene-environment interactions in cortical interneuron development and dysfunction: A review of preclinical studies. Neurotoxicology 2017; 58:120-129. [PMID: 27932026 PMCID: PMC5328258 DOI: 10.1016/j.neuro.2016.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 12/03/2016] [Accepted: 12/03/2016] [Indexed: 12/26/2022]
Abstract
Cortical interneurons (cINs) are a diverse group of locally projecting neurons essential to the organization and regulation of neural networks. Though they comprise only ∼20% of neurons in the neocortex, their dynamic modulation of cortical activity is requisite for normal cognition and underlies multiple aspects of learning and memory. While displaying significant morphological, molecular, and electrophysiological variability, cINs collectively function to maintain the excitatory-inhibitory balance in the cortex by dampening hyperexcitability and synchronizing activity of projection neurons, primarily through use of the inhibitory neurotransmitter gamma-aminobutyric acid (GABA). Disruption of the excitatory-inhibitory balance is a common pathophysiological feature of multiple seizure and neuropsychiatric disorders, including epilepsy, schizophrenia, and autism. While most studies have focused on genetic disruption of cIN development in these conditions, emerging evidence indicates that cIN development is exquisitely sensitive to teratogenic disruption. Here, we review key aspects of cIN development, including specification, migration, and integration into neural circuits. Additionally, we examine the mechanisms by which prenatal exposure to common chemical and environmental agents disrupt these events in preclinical models. Understanding how genetic and environmental factors interact to disrupt cIN development and function has tremendous potential to advance prevention and treatment of prevalent seizure and neuropsychiatric illnesses.
Collapse
Affiliation(s)
- Lydia J Ansen-Wilson
- Department of Comparative Biosciences School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA; Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA.
| | - Robert J Lipinski
- Department of Comparative Biosciences School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA; Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA; Molecular and Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, 1010B McArdle Building, 1400 University Avenue, Madison, WI, 53706, USA.
| |
Collapse
|
46
|
Sun Y, Paşca SP, Portmann T, Goold C, Worringer KA, Guan W, Chan KC, Gai H, Vogt D, Chen YJJ, Mao R, Chan K, Rubenstein JL, Madison DV, Hallmayer J, Froehlich-Santino WM, Bernstein JA, Dolmetsch RE. A deleterious Nav1.1 mutation selectively impairs telencephalic inhibitory neurons derived from Dravet Syndrome patients. eLife 2016; 5. [PMID: 27458797 PMCID: PMC4961470 DOI: 10.7554/elife.13073] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 06/30/2016] [Indexed: 12/12/2022] Open
Abstract
Dravet Syndrome is an intractable form of childhood epilepsy associated with deleterious mutations in SCN1A, the gene encoding neuronal sodium channel Nav1.1. Earlier studies using human induced pluripotent stem cells (iPSCs) have produced mixed results regarding the importance of Nav1.1 in human inhibitory versus excitatory neurons. We studied a Nav1.1 mutation (p.S1328P) identified in a pair of twins with Dravet Syndrome and generated iPSC-derived neurons from these patients. Characterization of the mutant channel revealed a decrease in current amplitude and hypersensitivity to steady-state inactivation. We then differentiated Dravet-Syndrome and control iPSCs into telencephalic excitatory neurons or medial ganglionic eminence (MGE)-like inhibitory neurons. Dravet inhibitory neurons showed deficits in sodium currents and action potential firing, which were rescued by a Nav1.1 transgene, whereas Dravet excitatory neurons were normal. Our study identifies biophysical impairments underlying a deleterious Nav1.1 mutation and supports the hypothesis that Dravet Syndrome arises from defective inhibitory neurons. DOI:http://dx.doi.org/10.7554/eLife.13073.001
Collapse
Affiliation(s)
- Yishan Sun
- Novartis Institutes for BioMedical Research, Cambridge, United States.,Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| | - Sergiu P Paşca
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States.,Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, United States
| | - Thomas Portmann
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| | - Carleton Goold
- Novartis Institutes for BioMedical Research, Cambridge, United States.,Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| | | | - Wendy Guan
- Novartis Institutes for BioMedical Research, Cambridge, United States
| | - Karen C Chan
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| | - Hui Gai
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| | - Daniel Vogt
- Department of Psychiatry, University of California, San Francisco, San Francisco, United States
| | - Ying-Jiun J Chen
- Department of Psychiatry, University of California, San Francisco, San Francisco, United States
| | - Rong Mao
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| | - Karrie Chan
- Novartis Institutes for BioMedical Research, Cambridge, United States
| | - John Lr Rubenstein
- Department of Psychiatry, University of California, San Francisco, San Francisco, United States
| | - Daniel V Madison
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| | - Joachim Hallmayer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, United States
| | - Wendy M Froehlich-Santino
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, United States
| | - Jonathan A Bernstein
- Department of Pediatrics, Division of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Ricardo E Dolmetsch
- Novartis Institutes for BioMedical Research, Cambridge, United States.,Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
47
|
Asmus SE, Raghanti MA, Beyerle ER, Fleming-Beattie JC, Hawkins SM, McKernan CM, Rauh NA. Tyrosine hydroxylase-producing neurons in the human cerebral cortex do not colocalize with calcium-binding proteins or the serotonin 3A receptor. J Chem Neuroanat 2016; 78:1-9. [PMID: 27448941 DOI: 10.1016/j.jchemneu.2016.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 07/14/2016] [Accepted: 07/15/2016] [Indexed: 11/25/2022]
Abstract
Interneurons of the cerebral cortex play a significant role in cortical information processing and are of clinical interest due to their involvement in neurological disorders. In the human neocortex, three subsets of interneurons can be identified based on the production of the calcium-binding proteins parvalbumin, calretinin or calbindin. A subset of interneurons in the mouse cortex expresses the serotonin 3A receptor (5-HT3AR). Previous work in humans has also demonstrated the presence of a subgroup of cortical neurons that produces the catecholaminergic enzyme tyrosine hydroxylase (TH). Many TH-producing cells in the rat cortex coexpress calretinin and are adjacent to blood vessels. However, little is known about the phenotype of these TH interneurons in humans. Here we immunohistochemically examined the coexpression of TH with parvalbumin, calretinin, calbindin or 5-HT3AR in human Brodmann's areas 10 and 24, cortical regions with high densities of TH-containing neurons. Colocalization of TH with these calcium-binding proteins and with 5-HT3AR was not detected in either area. Cortical TH cells were rarely apposed to blood vessels, denoted by immunolabeling for the gliovascular marker aquaporin-4. Our results suggest that the TH-immunoreactive cells in the human cortex do not overlap with any known neurochemically-defined subsets of interneurons and provide further evidence of differences in the phenotype of these cells across species.
Collapse
Affiliation(s)
- Stephen E Asmus
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA.
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Eric R Beyerle
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA
| | - Julia C Fleming-Beattie
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA
| | - Sarah M Hawkins
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA
| | - Courtney M McKernan
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA
| | - Nicholas A Rauh
- Biochemistry and Molecular Biology and Biology Programs, Centre College, Danville, KY 40422, USA
| |
Collapse
|
48
|
Larimer P, Spatazza J, Espinosa JS, Tang Y, Kaneko M, Hasenstaub AR, Stryker MP, Alvarez-Buylla A. Caudal Ganglionic Eminence Precursor Transplants Disperse and Integrate as Lineage-Specific Interneurons but Do Not Induce Cortical Plasticity. Cell Rep 2016; 16:1391-1404. [PMID: 27425623 DOI: 10.1016/j.celrep.2016.06.071] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/04/2016] [Accepted: 06/16/2016] [Indexed: 10/21/2022] Open
Abstract
The maturation of inhibitory GABAergic cortical circuits regulates experience-dependent plasticity. We recently showed that the heterochronic transplantation of parvalbumin (PV) or somatostatin (SST) interneurons from the medial ganglionic eminence (MGE) reactivates ocular dominance plasticity (ODP) in the postnatal mouse visual cortex. Might other types of interneurons similarly induce cortical plasticity? Here, we establish that caudal ganglionic eminence (CGE)-derived interneurons, when transplanted into the visual cortex of neonatal mice, migrate extensively in the host brain and acquire laminar distribution, marker expression, electrophysiological properties, and visual response properties like those of host CGE interneurons. Although transplants from the anatomical CGE do induce ODP, we found that this plasticity reactivation is mediated by a small fraction of MGE-derived cells contained in the transplant. These findings demonstrate that transplanted CGE cells can successfully engraft into the postnatal mouse brain and confirm the unique role of MGE lineage neurons in the induction of ODP.
Collapse
Affiliation(s)
- Phillip Larimer
- Center for Integrative Neuroscience, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA; Coleman Memorial Laboratory, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Julien Spatazza
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Juan Sebastian Espinosa
- Center for Integrative Neuroscience, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yunshuo Tang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Megumi Kaneko
- Center for Integrative Neuroscience, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Andrea R Hasenstaub
- Center for Integrative Neuroscience, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA; Coleman Memorial Laboratory, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael P Stryker
- Center for Integrative Neuroscience, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
49
|
Evidence That the Laminar Fate of LGE/CGE-Derived Neocortical Interneurons Is Dependent on Their Progenitor Domains. J Neurosci 2016; 36:2044-56. [PMID: 26865626 DOI: 10.1523/jneurosci.3550-15.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Neocortical interneurons show tremendous diversity in terms of their neurochemical marker expressions, morphology, electrophysiological properties, and laminar fate. Allocation of interneurons to their appropriate regions and layers in the neocortex is thought to play important roles for the emergence of higher functions of the neocortex. Neocortical interneurons mainly originate from the medial ganglionic eminence (MGE) and the caudal ganglionic eminence (CGE). The diversity and the laminar fate of MGE-derived interneurons depend on the location of their birth and birthdate, respectively. However, this relationship does not hold for CGE-derived interneurons. Here, using the method of in utero electroporation, which causes arbitrary occurrence of labeled progenitor domains, we tracked all descendants of the lateral ganglionic eminence (LGE)/CGE progenitors in mice. We provide evidence that neocortical interneurons with distinct laminar fate originate from distinct progenitor domains within the LGE/CGE. We find layer I interneurons are predominantly labeled in a set of animals, whereas other upper layer neurons are predominantly labeled in another set. We also find distinct subcortical structures labeled between the two sets. Further, interneurons labeled in layer I show distinct neurochemical properties from those in other layers. Together, these results suggest that the laminar fate of LGE/CGE-derived interneurons depends on their spatial origin. SIGNIFICANCE STATEMENT Diverse types of neocortical interneurons have distinct laminar fate, neurochemical marker expression, morphology, and electrophysiological properties. Although the specifications and laminar fate of medial ganglionic eminence-derived neocortical interneurons depend on their location of embryonic origin and birthdate, no similar causality of lateral/caudal ganglionic eminence (LGE/CGE)-derived neocortical interneurons is known. Here, we performed in utero electroporation on mouse LGE/CGE and found two groups of animals, one with preferential labeling of layer I and the other with preferential labeling of other layers. Interneurons labeled in these two groups show distinct neurochemical properties and morphologies and are associated with labeling of distinct subcortical structures. These findings suggest that the laminar fate of LGE/CGE-derived neocortical interneurons depends on their spatial origin.
Collapse
|
50
|
A New Mechanism of Receptor Targeting by Interaction between Two Classes of Ligand-Gated Ion Channels. J Neurosci 2016; 36:1456-70. [PMID: 26843630 DOI: 10.1523/jneurosci.2390-15.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED The 5-HT3 receptors are serotonin-gated ion channels that physically couple with purinergic P2X2 receptors to trigger a functional cross-inhibition leading to reciprocal channel occlusion. Although this functional receptor-receptor coupling seems to serve a modulatory role on both channels, this might not be its main physiological purpose. Using primary cultures of rat hippocampal neurons as a quantitative model of polarized targeting, we show here a novel function for this interaction. In this model, 5-HT3A receptors did not exhibit by themselves the capability of distal targeting in dendrites and axons but required the presence of P2X2R for their proper subcellular localization. 5-HT3AR distal targeting occurred with a delayed time course and exhibited a neuron phenotype dependency. In the subpopulation of neurons expressing endogenous P2X2R, 5-HT3AR distal neuritic localization correlated with P2X2R expression and could be selectively inhibited by P2X2R RNA interference. Cotransfection of both receptors revealed a specific colocalization, cotrafficking in common surface clusters, and the axonal rerouting of 5-HT3AR. The physical association between the two receptors was dependent on the second intracellular loop of the 5-HT3A subunit, but not on the P2X2R C-terminal tail that triggers the functional cross-inhibition with the 5-HT3AR. Together, these data establish that 5-HT3AR distal targeting in axons and dendrites primarily depends on P2X2R expression. Because several P2XR have now been shown to functionally interact with several other members of the 4-TMD family of receptor channels, we propose to reconsider the real functional role for this receptor family, as trafficking partner proteins dynamically involved in other receptors targeting. SIGNIFICANCE STATEMENT So far, receptor targeting mechanisms were found to involve intracellular partner proteins or supramolecular complexes that couple receptors to cytoskeletal elements and recruit them into cargo vesicles. In this paper, we describe a new trafficking mechanism for the neuronal serotonin 5-HT3A ionotropic channel receptor, in which the role of routing partner is endowed by a functionally interacting purinergic receptor: the P2X2 receptor. This work not only unveils the mechanism by which 5-HT3 receptors can reach their axonal localization required for the control of neurotransmitter release, but also suggests that, in addition to their modulatory role, the family of P2X receptors could have a previously undescribed functional role of trafficking partner proteins dynamically involved in the targeting of other receptors.
Collapse
|