1
|
Guo Z, Xiao S, Sun S, Su T, Tang X, Chen G, Chen P, Chen R, Chen C, Gong J, Yang Z, Huang L, Jia Y, Wang Y. Neural Activity Alterations and Their Association With Neurotransmitter and Genetic Profiles in Schizophrenia: Evidence From Clinical Patients and Unaffected Relatives. CNS Neurosci Ther 2025; 31:e70218. [PMID: 39924342 PMCID: PMC11807726 DOI: 10.1111/cns.70218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/11/2024] [Accepted: 01/03/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND The pattern of abnormal resting-state brain function has been documented in schizophrenia (SCZ). However, as of yet, it remains unclear whether this pattern is of genetic predisposition or related to the illness itself. METHODS A systematical meta-analysis was performed to identify resting-state functional differences in probands and their high-risk first-degree relatives of schizophrenia (FDRs-SCZ) using Seed-based d Mapping software. Subsequently, spatial associations between postmortem gene expression and neurotransmitters distribution data and neural activity alterations were conducted to uncover neural mechanisms underlaying FDRs-SCZ and SCZ from a multidimensional perspective. RESULTS A total of 13 studies comprising 503 FDRs-SCZ and 605 healthy controls (HCs) and 129 studies comprising 6506 patients with SCZ and 6982 HCs were included. Compared to HCs, FDRs-SCZ displayed increased spontaneous functional activity in the bilateral anterior cingulate cortex/medial prefrontal cortex (ACC/mPFC); patients with SCZ showed decreased spontaneous functional activity in the bilateral ACC/mPFC, bilateral postcentral gyrus, and right middle temporal gyrus as well as increased spontaneous functional activity in the bilateral striatum. The altered functional activity in FDRs-SCZ and SCZ shared similar spatial associations with genes enriched in potassium ion transmembrane transport, channel activity, and complex. The FDRs-SCZ and SCZ-related brain functional patterns were additionally associated with dopaminergic, serotonergic, and cholinergic neurotransmitter distribution. CONCLUSIONS SCZ-related resting-state functional, neuroimaging transcriptomes, and neurotransmitters abnormalities may exist in high-risk unaffected FDRs-SCZ, rather than just in overt SCZ. The study extended the evidence that altered brain function, along with their spatial correlations to genetics and neurotransmitter systems, may associate with genetic vulnerability for SCZ.
Collapse
Affiliation(s)
- Zixuan Guo
- Medical Imaging CenterFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
| | - Shu Xiao
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
- Department of Medical ImagingThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouChina
| | - Shilin Sun
- Medical Imaging CenterFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
| | - Ting Su
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
- Department of RadiologyThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Xinyue Tang
- Medical Imaging CenterFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
| | - Guanmao Chen
- Medical Imaging CenterFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
| | - Pan Chen
- Medical Imaging CenterFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
| | - Ruoyi Chen
- Medical Imaging CenterFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
| | - Chao Chen
- Medical Imaging CenterFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
| | - Jiaying Gong
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
- Department of RadiologySix Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Zibin Yang
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
- Department of Medical ImagingThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouChina
| | - Li Huang
- Medical Imaging CenterFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
| | - Yanbin Jia
- Department of PsychiatryFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Ying Wang
- Medical Imaging CenterFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
- Institute of Molecular and Functional ImagingJinan UniversityGuangzhouChina
| |
Collapse
|
2
|
Fournier LA, Phadke RA, Salgado M, Brack A, Nocon JC, Bolshakova S, Grant JR, Padró Luna NM, Sen K, Cruz-Martín A. Overexpression of the schizophrenia risk gene C4 in PV cells drives sex-dependent behavioral deficits and circuit dysfunction. iScience 2024; 27:110800. [PMID: 39310747 PMCID: PMC11416532 DOI: 10.1016/j.isci.2024.110800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Fast-spiking parvalbumin (PV)-positive cells are key players in orchestrating pyramidal neuron activity, and their dysfunction is consistently observed in myriad brain diseases. To understand how immune complement pathway dysregulation in PV cells drives disease pathogenesis, we have developed a transgenic line that permits cell-type specific overexpression of the schizophrenia-associated C4 gene. We found that overexpression of mouse C4 (mC4) in PV cells causes sex-specific alterations in anxiety-like behavior and deficits in synaptic connectivity and excitability of PFC PV cells. Using a computational model, we demonstrated that these microcircuit deficits led to hyperactivity and disrupted neural communication. Finally, pan-neuronal overexpression of mC4 failed to evoke the same deficits in behavior as PV-specific mC4 overexpression, suggesting that perturbations of this neuroimmune gene in fast-spiking neurons are especially detrimental to circuits associated with anxiety-like behavior. Together, these results provide a causative link between C4 and the vulnerability of PV cells in brain disease.
Collapse
Affiliation(s)
- Luke A. Fournier
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Rhushikesh A. Phadke
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Maria Salgado
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Alison Brack
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Jian Carlo Nocon
- Neurophotonics Center, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Hearing Research Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sonia Bolshakova
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics MS Program, Boston University, Boston, MA, USA
| | - Jaylyn R. Grant
- Biological Sciences, Eastern Illinois University, Charleston, IL, USA
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, MA, USA
| | - Nicole M. Padró Luna
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, MA, USA
- Biology Department, College of Natural Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, USA
| | - Kamal Sen
- Neurophotonics Center, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Hearing Research Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Alberto Cruz-Martín
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- NeuroTechnology Center (NTC), University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
3
|
Miyazaki Y, Otsuka T, Yamagata Y, Endo T, Sanbo M, Sano H, Kobayashi K, Inahashi H, Kornau HC, Schmitz D, Prüss H, Meijer D, Hirabayashi M, Fukata Y, Fukata M. Oligodendrocyte-derived LGI3 and its receptor ADAM23 organize juxtaparanodal Kv1 channel clustering for short-term synaptic plasticity. Cell Rep 2024; 43:113634. [PMID: 38194969 PMCID: PMC10828548 DOI: 10.1016/j.celrep.2023.113634] [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: 09/09/2022] [Revised: 10/31/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Neurodevelopmental disorders, such as intellectual disability (ID), epilepsy, and autism, involve altered synaptic transmission and plasticity. Functional characterization of their associated genes is vital for understanding physio-pathological brain functions. LGI3 is a recently recognized ID-associated gene encoding a secretory protein related to an epilepsy-gene product, LGI1. Here, we find that LGI3 is uniquely secreted from oligodendrocytes in the brain and enriched at juxtaparanodes of myelinated axons, forming nanoscale subclusters. Proteomic analysis using epitope-tagged Lgi3 knockin mice shows that LGI3 uses ADAM23 as a receptor and selectively co-assembles with Kv1 channels. A lack of Lgi3 in mice disrupts juxtaparanodal clustering of ADAM23 and Kv1 channels and suppresses Kv1-channel-mediated short-term synaptic plasticity. Collectively, this study identifies an extracellular organizer of juxtaparanodal Kv1 channel clustering for finely tuned synaptic transmission. Given the defective secretion of the LGI3 missense variant, we propose a molecular pathway, the juxtaparanodal LGI3-ADAM23-Kv1 channel, for understanding neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yuri Miyazaki
- Division of Neuropharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Takeshi Otsuka
- Section of Cellular Electrophysiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan
| | - Yoko Yamagata
- Section of Multilayer Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | | | - Makoto Sanbo
- Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Hiromi Sano
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Kenta Kobayashi
- Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan; Section of Viral Vector Development, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | - Hiroki Inahashi
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Hans-Christian Kornau
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Neuroscience Research Center (NWFZ), Cluster NeuroCure, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Neuroscience Research Center (NWFZ), Cluster NeuroCure, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Helmholtz Innovation Lab BaoBab (Brain Antibody-omics and B-cell Lab), Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dies Meijer
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK; Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK
| | - Masumi Hirabayashi
- Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan; Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Yuko Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Division of Molecular and Cellular Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Masaki Fukata
- Division of Neuropharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan.
| |
Collapse
|
4
|
Sylte OC, Muysers H, Chen HL, Bartos M, Sauer JF. Neuronal tuning to threat exposure remains stable in the mouse prefrontal cortex over multiple days. PLoS Biol 2024; 22:e3002475. [PMID: 38206890 PMCID: PMC10783789 DOI: 10.1371/journal.pbio.3002475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 12/19/2023] [Indexed: 01/13/2024] Open
Abstract
Intense threat elicits action in the form of active and passive coping. The medial prefrontal cortex (mPFC) executes top-level control over the selection of threat coping strategies, but the dynamics of mPFC activity upon continuing threat encounters remain unexplored. Here, we used 1-photon calcium imaging in mice to probe the activity of prefrontal pyramidal cells during repeated exposure to intense threat in a tail suspension (TS) paradigm. A subset of prefrontal neurons displayed selective activation during TS, which was stably maintained over days. During threat, neurons showed specific tuning to active or passive coping. These responses were unrelated to general motion tuning and persisted over days. Moreover, the neural manifold traversed by low-dimensional population activity remained stable over subsequent days of TS exposure and was preserved across individuals. These data thus reveal a specific, temporally, and interindividually conserved repertoire of prefrontal tuning to behavioral responses under threat.
Collapse
Affiliation(s)
- Ole Christian Sylte
- University of Freiburg, Medical Faculty, Institute of Physiology I, Freiburg, Germany
- University of Freiburg, Faculty of Biology, Freiburg, Germany
| | - Hannah Muysers
- University of Freiburg, Medical Faculty, Institute of Physiology I, Freiburg, Germany
- University of Freiburg, Faculty of Biology, Freiburg, Germany
| | - Hung-Ling Chen
- University of Freiburg, Medical Faculty, Institute of Physiology I, Freiburg, Germany
| | - Marlene Bartos
- University of Freiburg, Medical Faculty, Institute of Physiology I, Freiburg, Germany
| | - Jonas-Frederic Sauer
- University of Freiburg, Medical Faculty, Institute of Physiology I, Freiburg, Germany
| |
Collapse
|
5
|
Martínez-Aguirre C, Márquez LA, Santiago-Castañeda CL, Carmona-Cruz F, Nuñez-Lumbreras MDLA, Martínez-Rojas VA, Alonso-Vanegas M, Aguado-Carrillo G, Gómez-Víquez NL, Galván EJ, Cuéllar-Herrera M, Rocha L. Cannabidiol Modifies the Glutamate Over-Release in Brain Tissue of Patients and Rats with Epilepsy: A Pilot Study. Biomedicines 2023; 11:3237. [PMID: 38137458 PMCID: PMC10741033 DOI: 10.3390/biomedicines11123237] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Drug-resistant epilepsy (DRE) is associated with high extracellular levels of glutamate. Studies support the idea that cannabidiol (CBD) decreases glutamate over-release. This study focused on investigating whether CBD reduces the evoked glutamate release in cortical synaptic terminals obtained from patients with DRE as well as in a preclinical model of epilepsy. Synaptic terminals (synaptosomes) were obtained from the epileptic neocortex of patients with drug-resistant temporal lobe epilepsy (DR-TLE, n = 10) or drug-resistant extratemporal lobe epilepsy (DR-ETLE, n = 10) submitted to epilepsy surgery. Synaptosomes highly purified by Percoll-sucrose density gradient were characterized by confocal microscopy and Western blot. Synaptosomes were used to estimate the high KCl (33 mM)-evoked glutamate release in the presence of CBD at different concentrations. Our results revealed responsive tissue obtained from seven patients with DR-TLE and seven patients with DR-ETLE. Responsive tissue showed lower glutamate release (p < 0.05) when incubated with CBD at low concentrations (less than 100 µM) but not at higher concentrations. Tissue that was non-responsive to CBD (DR-TLE, n = 3 and DR-ELTE, n = 3) showed high glutamate release despite CBD exposure at different concentrations. Simultaneously, a block of the human epileptic neocortex was used to determine its viability through whole-cell and extracellular electrophysiological recordings. The electrophysiological evaluations supported that the responsive and non-responsive human epileptic neocortices used in the present study exhibited proper neuronal viability and stability to acquire electrophysiological responses. We also investigated whether the subchronic administration of CBD could reduce glutamate over-release in a preclinical model of temporal lobe epilepsy. Administration of CBD (200 mg/kg, p.o. every 24 h for 7 days) to rats with lithium-pilocarpine-evoked spontaneous recurrent seizures reduced glutamate over-release in the hippocampus. The present study revealed that acute exposure to low concentrations of CBD can reduce the glutamate over-release in synaptic terminals obtained from some patients with DRE. This effect is also evident when applied subchronically in rats with spontaneous recurrent seizures. An important finding was the identification of a group of patients that were non-responsive to CBD effects. Future studies are essential to identify biomarkers of responsiveness to CBD to control DRE.
Collapse
Affiliation(s)
- Christopher Martínez-Aguirre
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Luis Alfredo Márquez
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Cindy Lizbeth Santiago-Castañeda
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Francia Carmona-Cruz
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Maria de los Angeles Nuñez-Lumbreras
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Vladimir A. Martínez-Rojas
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
- Center for Research on Aging, Center for Research and Advanced Studies, Mexico City 14330, Mexico
| | - Mario Alonso-Vanegas
- International Center for Epilepsy Surgery, HMG-Coyoacán Hospital, Mexico City 04380, Mexico;
| | - Gustavo Aguado-Carrillo
- Clinic of Epilepsy, General Hospital of México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Norma L. Gómez-Víquez
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Emilio J. Galván
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
- Center for Research on Aging, Center for Research and Advanced Studies, Mexico City 14330, Mexico
| | - Manola Cuéllar-Herrera
- Clinic of Epilepsy, General Hospital of México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Luisa Rocha
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| |
Collapse
|
6
|
Reva M, Rössert C, Arnaudon A, Damart T, Mandge D, Tuncel A, Ramaswamy S, Markram H, Van Geit W. A universal workflow for creation, validation, and generalization of detailed neuronal models. PATTERNS (NEW YORK, N.Y.) 2023; 4:100855. [PMID: 38035193 PMCID: PMC10682753 DOI: 10.1016/j.patter.2023.100855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/24/2023] [Accepted: 09/12/2023] [Indexed: 12/02/2023]
Abstract
Detailed single-neuron modeling is widely used to study neuronal functions. While cellular and functional diversity across the mammalian cortex is vast, most of the available computational tools focus on a limited set of specific features characteristic of a single neuron. Here, we present a generalized automated workflow for the creation of robust electrical models and illustrate its performance by building cell models for the rat somatosensory cortex. Each model is based on a 3D morphological reconstruction and a set of ionic mechanisms. We use an evolutionary algorithm to optimize neuronal parameters to match the electrophysiological features extracted from experimental data. Then we validate the optimized models against additional stimuli and assess their generalizability on a population of similar morphologies. Compared to the state-of-the-art canonical models, our models show 5-fold improved generalizability. This versatile approach can be used to build robust models of any neuronal type.
Collapse
Affiliation(s)
- Maria Reva
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Christian Rössert
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Alexis Arnaudon
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Tanguy Damart
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Darshan Mandge
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Anıl Tuncel
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Srikanth Ramaswamy
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| | - Henry Markram
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
- Laboratory of Neural Microcircuitry (LNMC), Brain Mind Institute, School of Life Sciences, École polytechnique fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Werner Van Geit
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Campus Biotech, 1202 Geneva, Switzerland
| |
Collapse
|
7
|
Gebicke-Haerter PJ. The computational power of the human brain. Front Cell Neurosci 2023; 17:1220030. [PMID: 37608987 PMCID: PMC10441807 DOI: 10.3389/fncel.2023.1220030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/05/2023] [Indexed: 08/24/2023] Open
Abstract
At the end of the 20th century, analog systems in computer science have been widely replaced by digital systems due to their higher computing power. Nevertheless, the question keeps being intriguing until now: is the brain analog or digital? Initially, the latter has been favored, considering it as a Turing machine that works like a digital computer. However, more recently, digital and analog processes have been combined to implant human behavior in robots, endowing them with artificial intelligence (AI). Therefore, we think it is timely to compare mathematical models with the biology of computation in the brain. To this end, digital and analog processes clearly identified in cellular and molecular interactions in the Central Nervous System are highlighted. But above that, we try to pinpoint reasons distinguishing in silico computation from salient features of biological computation. First, genuinely analog information processing has been observed in electrical synapses and through gap junctions, the latter both in neurons and astrocytes. Apparently opposed to that, neuronal action potentials (APs) or spikes represent clearly digital events, like the yes/no or 1/0 of a Turing machine. However, spikes are rarely uniform, but can vary in amplitude and widths, which has significant, differential effects on transmitter release at the presynaptic terminal, where notwithstanding the quantal (vesicular) release itself is digital. Conversely, at the dendritic site of the postsynaptic neuron, there are numerous analog events of computation. Moreover, synaptic transmission of information is not only neuronal, but heavily influenced by astrocytes tightly ensheathing the majority of synapses in brain (tripartite synapse). At least at this point, LTP and LTD modifying synaptic plasticity and believed to induce short and long-term memory processes including consolidation (equivalent to RAM and ROM in electronic devices) have to be discussed. The present knowledge of how the brain stores and retrieves memories includes a variety of options (e.g., neuronal network oscillations, engram cells, astrocytic syncytium). Also epigenetic features play crucial roles in memory formation and its consolidation, which necessarily guides to molecular events like gene transcription and translation. In conclusion, brain computation is not only digital or analog, or a combination of both, but encompasses features in parallel, and of higher orders of complexity.
Collapse
Affiliation(s)
- Peter J. Gebicke-Haerter
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
8
|
Nimitvilai-Roberts S, Gioia D, Lopez MF, Glaser CM, Woodward JJ. Chronic intermittent ethanol exposure differentially alters the excitability of neurons in the orbitofrontal cortex and basolateral amygdala that project to the dorsal striatum. Neuropharmacology 2023; 228:109463. [PMID: 36792030 PMCID: PMC10006395 DOI: 10.1016/j.neuropharm.2023.109463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023]
Abstract
Alcohol use disorder is associated with altered neuron function including those in orbitofrontal cortex (OFC) and basolateral amygdala (BLA) that send glutamatergic inputs to areas of the dorsal striatum (DS) that mediate goal and habit directed actions. Previous studies reported that chronic intermittent (CIE) exposure to ethanol alters the electrophysiological properties of OFC and BLA neurons, although projection targets for these neurons were not identified. In this study, we used male and female mice and recorded current-evoked spiking of retrobead labeled DS-projecting OFC and BLA neurons in the same animals following air or CIE treatment. DS-projecting OFC neurons were hyperexcitable 3- and 7-days following CIE exposure and spiking returned to control levels after 14 days of withdrawal. In contrast, firing was decreased in DS-projecting BLA neurons at 3-days withdrawal, increased at 7- and 14-days and returned to baseline at 28 days post-CIE. CIE exposure enhanced the amplitude and frequency of spontaneous excitatory postsynaptic currents (sEPSCs) of DS-projecting OFC neurons but had no effect on inhibitory postsynaptic currents (sIPSCs). In DS-projecting BLA neurons, the amplitude and frequency of sIPSCs was enhanced 3 days post-CIE with no change in sEPSCs while at 7-days post-withdrawal, sEPSC amplitude and frequency were increased and sIPSCs had returned to normal. Finally, in CIE-treated mice, acute ethanol no longer inhibited spike firing of DS-projecting OFC and BLA neurons. Overall, these results suggest that CIE-induced changes in the excitability of DS-projecting OFC and BLA neurons could underlie deficits in behavioral control often observed in alcohol-dependent individuals.
Collapse
Affiliation(s)
| | - Dominic Gioia
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Marcelo F Lopez
- Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Christina M Glaser
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
9
|
Tang W, Cory B, Lim KL, Fivaz M. The Mood Stabilizer Lithium Slows Down Synaptic Vesicle Cycling at Glutamatergic Synapses. Neuromolecular Med 2023; 25:125-135. [PMID: 36436129 DOI: 10.1007/s12017-022-08729-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/31/2022] [Indexed: 11/28/2022]
Abstract
Lithium is a mood stabilizer broadly used to prevent and treat symptoms of mania and depression in people with bipolar disorder (BD). Little is known, however, about its mode of action. Here, we analyzed the impact of lithium on synaptic vesicle (SV) cycling at presynaptic terminals releasing glutamate, a neurotransmitter previously implicated in BD and other neuropsychiatric conditions. We used the pHluorin-based synaptic tracer vGpH and a fully automated image processing pipeline to quantify the effect of lithium on both SV exocytosis and endocytosis in hippocampal neurons. We found that lithium selectively reduces SV exocytic rates during electrical stimulation, and markedly slows down SV recycling post-stimulation. Analysis of single-bouton responses revealed the existence of functionally distinct excitatory synapses with varying sensitivity to lithium-some terminals show responses similar to untreated cells, while others are markedly impaired in their ability to recycle SVs. While the cause of this heterogeneity is unclear, these data indicate that lithium interacts with the SV machinery and influences glutamate release in a large fraction of excitatory synapses. Together, our findings show that lithium down modulates SV cycling, an effect consistent with clinical reports indicating hyperactivation of glutamate neurotransmission in BD.
Collapse
Affiliation(s)
- Willcyn Tang
- Department of Research, Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
- Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Bradley Cory
- Stem Cell & Gene Editing Laboratory, Faculty of Science and Engineering, University of Greenwich, Kent, ME4 4TB, UK
| | - Kah-Leong Lim
- Department of Research, Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore.
- Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore.
- Department of Brain Sciences, Imperial College London, London, SW7 2AZ, UK.
| | - Marc Fivaz
- Stem Cell & Gene Editing Laboratory, Faculty of Science and Engineering, University of Greenwich, Kent, ME4 4TB, UK.
- reMYND NV, Bio-Incubator, Gaston Geenslaan 1, Heverlee, 3001, Leuven, Belgium.
| |
Collapse
|
10
|
Synaptic plasticity in Schizophrenia pathophysiology. IBRO Neurosci Rep 2023. [DOI: 10.1016/j.ibneur.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
11
|
Fischer M, Zimmerman A, Zhang E, Kolis J, Dickey A, Burdette MK, Chander P, Foulger SH, Brigman JL, Weick JP. Distribution and inflammatory cell response to intracranial delivery of radioluminescent Y2(SiO4)O:Ce particles. PLoS One 2023; 18:e0276819. [PMID: 36634053 PMCID: PMC9836305 DOI: 10.1371/journal.pone.0276819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 10/13/2022] [Indexed: 01/13/2023] Open
Abstract
Due to increasing advances in their manufacture and functionalization, nanoparticle-based systems have become a popular tool for in vivo drug delivery and biodetection. Recently, scintillating nanoparticles such as yttrium orthosilicate doped with cerium (Y2(SiO4)O:Ce) have come under study for their potential utility in optogenetic applications, as they emit photons upon low levels of stimulation from remote x-ray sources. The utility of such nanoparticles in vivo is hampered by rapid clearance from circulation by the mononuclear phagocytic system, which heavily restricts nanoparticle accumulation at target tissues. Local transcranial injection of nanoparticles may deliver scintillating nanoparticles to highly specific brain regions by circumventing the blood-brain barrier and avoiding phagocytic clearance. Few studies to date have examined the distribution and response to nanoparticles following localized delivery to cerebral cortex, a crucial step in understanding the therapeutic potential of nanoparticle-based biodetection in the brain. Following the synthesis and surface modification of these nanoparticles, two doses (1 and 3 mg/ml) were introduced into mouse secondary motor cortex (M2). This region was chosen as the site for RLP delivery, as it represents a common target for optogenetic manipulations of mouse behavior, and RLPs could eventually serve as an injectable x-ray inducible light delivery system. The spread of particles through the target tissue was assessed 24 hours, 72 hours, and 9 days post-injection. Y2(SiO4)O:Ce nanoparticles were found to be detectable in the brain for up to 9 days, initially diffusing through the tissue until 72 hours before achieving partial clearance by the final endpoint. Small transient increases in the presence of IBA-1+ microglia and GFAP+ astrocytic cell populations were detected near nanoparticle injection sites of both doses tested 24 hours after surgery. Taken together, these data provide evidence that Y2(SiO4)O:Ce nanoparticles coated with BSA can be injected directly into mouse cortex in vivo, where they persist for days and are broadly tolerated, such that they may be potentially utilized for remote x-ray activated stimulation and photon emission for optogenetic experiments in the near future.
Collapse
Affiliation(s)
- Máté Fischer
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
| | - Amber Zimmerman
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
| | - Eric Zhang
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina, United States of America
| | - Joseph Kolis
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina, United States of America
| | - Ashley Dickey
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina, United States of America
| | - Mary K. Burdette
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina, United States of America
| | - Praveen Chander
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
| | - Stephen H. Foulger
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina, United States of America
- Center for Optical Materials Science and Engineering Technologies, Clemson University, Clemson, South Carolina, United States of America
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Jonathan L. Brigman
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
- Center for Brain Recovery and Repair, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
| | - Jason P. Weick
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
- Center for Brain Recovery and Repair, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
| |
Collapse
|
12
|
Günther A, Hanganu-Opatz IL. Neuronal oscillations: early biomarkers of psychiatric disease? Front Behav Neurosci 2022; 16:1038981. [PMID: 36600993 PMCID: PMC9806131 DOI: 10.3389/fnbeh.2022.1038981] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/09/2022] [Indexed: 12/23/2022] Open
Abstract
Our understanding of the environmental and genetic factors contributing to the wide spectrum of neuropsychiatric disorders has significantly increased in recent years. Impairment of neuronal network activity during early development has been suggested as a contributor to the emergence of neuropsychiatric pathologies later in life. Still, the neurobiological substrates underlying these disorders remain yet to be fully understood and the lack of biomarkers for early diagnosis has impeded research into curative treatment options. Here, we briefly review current knowledge on potential biomarkers for emerging neuropsychiatric disease. Moreover, we summarize recent findings on aberrant activity patterns in the context of psychiatric disease, with a particular focus on their potential as early biomarkers of neuropathologies, an essential step towards pre-symptomatic diagnosis and, thus, early intervention.
Collapse
|
13
|
Zhang K, Liao P, Wen J, Hu Z. Synaptic plasticity in schizophrenia pathophysiology. IBRO Neurosci Rep 2022; 13:478-487. [PMID: 36590092 PMCID: PMC9795311 DOI: 10.1016/j.ibneur.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 10/21/2022] [Indexed: 11/05/2022] Open
Abstract
Schizophrenia is a severe neuropsychiatric syndrome with psychotic behavioral abnormalities and marked cognitive deficits. It is widely accepted that genetic and environmental factors contribute to the onset of schizophrenia. However, the etiology and pathology of the disease remain largely unexplored. Recently, the synaptopathology and the dysregulated synaptic plasticity and function have emerging as intriguing and prominent biological mechanisms of schizophrenia pathogenesis. Synaptic plasticity is the ability of neurons to change the strength of their connections in response to internal or external stimuli, which is essential for brain development and function, learning and memory, and vast majority of behavior responses relevant to psychiatric diseases including schizophrenia. Here, we reviewed molecular and cellular mechanisms of the multiple forms synaptic plasticity, and the functional regulations of schizophrenia-risk factors including disease susceptible genes and environmental alterations on synaptic plasticity and animal behavior. Recent genome-wide association studies have provided fruitful findings of hundreds of risk gene variances associated with schizophrenia, thus further clarifying the role of these disease-risk genes in synaptic transmission and plasticity will be beneficial to advance our understanding of schizophrenia pathology, as well as the molecular mechanism of synaptic plasticity.
Collapse
Affiliation(s)
- Kexuan Zhang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China
| | - Panlin Liao
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Jin Wen
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhonghua Hu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China,Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha 410008, Hunan, PR China,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha 410008, Hunan, PR China,Correspondence to: Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, Hunan, PR China.
| |
Collapse
|
14
|
Sauer JF, Bartos M. Disrupted-in-schizophrenia-1 is required for normal pyramidal cell-interneuron communication and assembly dynamics in the prefrontal cortex. eLife 2022; 11:79471. [PMID: 36239988 PMCID: PMC9566853 DOI: 10.7554/elife.79471] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
We interrogated prefrontal circuit function in mice lacking Disrupted-in-schizophrenia-1 (Disc1-mutant mice), a risk factor for psychiatric disorders. Single-unit recordings in awake mice revealed reduced average firing rates of fast-spiking interneurons (INTs), including optogenetically identified parvalbumin-positive cells, and a lower proportion of INTs phase-coupled to ongoing gamma oscillations. Moreover, we observed decreased spike transmission efficacy at local pyramidal cell (PYR)-INT connections in vivo, suggesting a reduced excitatory effect of local glutamatergic inputs as a potential mechanism of lower INT rates. On the network level, impaired INT function resulted in altered activation of PYR assemblies: While assembly activations defined as coactivations within 25 ms were observed equally often, the expression strength of individual assembly patterns was significantly higher in Disc1-mutant mice. Our data, thus, reveal a role of Disc1 in shaping the properties of prefrontal assembly patterns by setting INT responsiveness to glutamatergic drive.
Collapse
Affiliation(s)
- Jonas-Frederic Sauer
- Institute for Physiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marlene Bartos
- Institute for Physiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
15
|
Rabadan MA, De La Cruz ED, Rao SB, Chen Y, Gong C, Crabtree G, Xu B, Markx S, Gogos JA, Yuste R, Tomer R. An in vitro model of neuronal ensembles. Nat Commun 2022; 13:3340. [PMID: 35680927 PMCID: PMC9184643 DOI: 10.1038/s41467-022-31073-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/01/2022] [Indexed: 11/28/2022] Open
Abstract
Advances in 3D neuronal cultures, such as brain spheroids and organoids, are allowing unprecedented in vitro access to some of the molecular, cellular and developmental mechanisms underlying brain diseases. However, their efficacy in recapitulating brain network properties that encode brain function remains limited, thereby precluding development of effective in vitro models of complex brain disorders like schizophrenia. Here, we develop and characterize a Modular Neuronal Network (MoNNet) approach that recapitulates specific features of neuronal ensemble dynamics, segregated local-global network activities and a hierarchical modular organization. We utilized MoNNets for quantitative in vitro modelling of schizophrenia-related network dysfunctions caused by highly penetrant mutations in SETD1A and 22q11.2 risk loci. Furthermore, we demonstrate its utility for drug discovery by performing pharmacological rescue of alterations in neuronal ensembles stability and global network synchrony. MoNNets allow in vitro modelling of brain diseases for investigating the underlying neuronal network mechanisms and systematic drug discovery.
Collapse
Affiliation(s)
- M Angeles Rabadan
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | | | - Sneha B Rao
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY, USA
| | - Yannan Chen
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Cheng Gong
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Gregg Crabtree
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY, USA
| | - Bin Xu
- Department of Psychiatry, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Sander Markx
- Department of Psychiatry, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Joseph A Gogos
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY, USA
- Department of Physiology, Columbia University, New York, NY, USA
- Department of Neuroscience, Columbia University, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - Rafael Yuste
- Department of Biological Sciences, Columbia University, New York, NY, USA
- NeuroTechnology Center, Columbia University, New York, NY, USA
| | - Raju Tomer
- Department of Biological Sciences, Columbia University, New York, NY, USA.
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
- NeuroTechnology Center, Columbia University, New York, NY, USA.
| |
Collapse
|
16
|
Adeyelu T, Shrestha A, Adeniyi PA, Lee CC, Ogundele OM. CA1 Spike Timing is Impaired in the 129S Inbred Strain During Cognitive Tasks. Neuroscience 2022; 484:119-138. [PMID: 34800576 PMCID: PMC8844212 DOI: 10.1016/j.neuroscience.2021.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 01/16/2023]
Abstract
A spontaneous mutation of the disrupted in schizophrenia 1 (Disc1) gene is carried by the 129S inbred mouse strain. Truncated DISC1 protein in 129S mouse synapses impairs the scaffolding of excitatory postsynaptic receptors and leads to progressive spine dysgenesis. In contrast, C57BL/6 inbred mice carry the wild-type Disc1 gene and exhibit more typical cognitive performance in spatial exploration and executive behavioral tests. Because of the innate Disc1 mutation, adult 129S inbred mice exhibit the behavioral phenotypes of outbred B6 Disc1 knockdown (Disc1-/-) or Disc1-L-100P mutant strains. Recent studies in Disc1-/- and L-100P mice have shown that impaired excitation-driven interneuron activity and low hippocampal theta power underlie the behavioral phenotypes that resemble human depression and schizophrenia. The current study compared the firing rate and connectivity profile of putative neurons in the CA1 of freely behaving inbred 129S and B6 mice, which have mutant and wild-type Disc1 genes, respectively. In cognitive behavioral tests, 129S mice had lower exploration scores than B6 mice. Furthermore, the mean firing rate for 129S putative pyramidal (pyr) cells and interneurons (int) was significantly lower than that for B6 CA1 neurons sampled during similar tasks. Analysis of pyr/int connectivity revealed a significant delay in synaptic transmission for 129S putative pairs. Sampled 129S pyr/int pairs also had lower detectability index scores than B6 putative pairs. Therefore, the spontaneous Disc1 mutation in the 129S strain attenuates the firing of putative pyr CA1 neurons and impairs spike timing fidelity during cognitive tasks.
Collapse
Affiliation(s)
- Tolulope Adeyelu
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine. Baton Rouge, LA70803, Louisiana
| | - Amita Shrestha
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine. Baton Rouge, LA70803, Louisiana
| | - Philip A. Adeniyi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine. Baton Rouge, LA70803, Louisiana
| | - Charles C. Lee
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine. Baton Rouge, LA70803, Louisiana
| | - Olalekan M. Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine. Baton Rouge, LA70803, Louisiana
| |
Collapse
|
17
|
Zbili M, Rama S, Benitez MJ, Fronzaroli-Molinieres L, Bialowas A, Boumedine-Guignon N, Garrido JJ, Debanne D. Homeostatic regulation of axonal Kv1.1 channels accounts for both synaptic and intrinsic modifications in the hippocampal CA3 circuit. Proc Natl Acad Sci U S A 2021; 118:e2110601118. [PMID: 34799447 PMCID: PMC8617510 DOI: 10.1073/pnas.2110601118] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/18/2022] Open
Abstract
Homeostatic plasticity of intrinsic excitability goes hand in hand with homeostatic plasticity of synaptic transmission. However, the mechanisms linking the two forms of homeostatic regulation have not been identified so far. Using electrophysiological, imaging, and immunohistochemical techniques, we show here that blockade of excitatory synaptic receptors for 2 to 3 d induces an up-regulation of both synaptic transmission at CA3-CA3 connections and intrinsic excitability of CA3 pyramidal neurons. Intrinsic plasticity was found to be mediated by a reduction of Kv1.1 channel density at the axon initial segment. In activity-deprived circuits, CA3-CA3 synapses were found to express a high release probability, an insensitivity to dendrotoxin, and a lack of depolarization-induced presynaptic facilitation, indicating a reduction in presynaptic Kv1.1 function. Further support for the down-regulation of axonal Kv1.1 channels in activity-deprived neurons was the broadening of action potentials measured in the axon. We conclude that regulation of the axonal Kv1.1 channel constitutes a major mechanism linking intrinsic excitability and synaptic strength that accounts for the functional synergy existing between homeostatic regulation of intrinsic excitability and synaptic transmission.
Collapse
Affiliation(s)
- Mickaël Zbili
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Sylvain Rama
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Maria-José Benitez
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid 28002, Spain
- Departamento de Química Física Aplicada, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Laure Fronzaroli-Molinieres
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Andrzej Bialowas
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Norah Boumedine-Guignon
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Juan José Garrido
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid 28002, Spain
| | - Dominique Debanne
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France;
| |
Collapse
|
18
|
Mizusaki BEP, O'Donnell C. Neural circuit function redundancy in brain disorders. Curr Opin Neurobiol 2021; 70:74-80. [PMID: 34416675 PMCID: PMC8694099 DOI: 10.1016/j.conb.2021.07.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/17/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023]
Abstract
Redundancy is a ubiquitous property of the nervous system. This means that vastly different configurations of cellular and synaptic components can enable the same neural circuit functions. However, until recently, very little brain disorder research has considered the implications of this characteristic when designing experiments or interpreting data. Here, we first summarise the evidence for redundancy in healthy brains, explaining redundancy and three related sub-concepts: sloppiness, dependencies and multiple solutions. We then lay out key implications for brain disorder research, covering recent examples of redundancy effects in experimental studies on psychiatric disorders. Finally, we give predictions for future experiments based on these concepts.
Collapse
Affiliation(s)
- Beatriz E P Mizusaki
- Computational Neuroscience Unit, School of Computer Science, Electrical and Electronic Engineering, and Engineering Mathematics, University of Bristol, BS8 1UB, United Kingdom
| | - Cian O'Donnell
- Computational Neuroscience Unit, School of Computer Science, Electrical and Electronic Engineering, and Engineering Mathematics, University of Bristol, BS8 1UB, United Kingdom.
| |
Collapse
|
19
|
Genestine M, Ambriz D, Crabtree GW, Dummer P, Molotkova A, Quintero M, Mela A, Biswas S, Feng H, Zhang C, Canoll P, Hargus G, Agalliu D, Gogos JA, Au E. Vascular-derived SPARC and SerpinE1 regulate interneuron tangential migration and accelerate functional maturation of human stem cell-derived interneurons. eLife 2021; 10:e56063. [PMID: 33904394 PMCID: PMC8099424 DOI: 10.7554/elife.56063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Cortical interneurons establish inhibitory microcircuits throughout the neocortex and their dysfunction has been implicated in epilepsy and neuropsychiatric diseases. Developmentally, interneurons migrate from a distal progenitor domain in order to populate the neocortex - a process that occurs at a slower rate in humans than in mice. In this study, we sought to identify factors that regulate the rate of interneuron maturation across the two species. Using embryonic mouse development as a model system, we found that the process of initiating interneuron migration is regulated by blood vessels of the medial ganglionic eminence (MGE), an interneuron progenitor domain. We identified two endothelial cell-derived paracrine factors, SPARC and SerpinE1, that enhance interneuron migration in mouse MGE explants and organotypic cultures. Moreover, pre-treatment of human stem cell-derived interneurons (hSC-interneurons) with SPARC and SerpinE1 prior to transplantation into neonatal mouse cortex enhanced their migration and morphological elaboration in the host cortex. Further, SPARC and SerpinE1-treated hSC-interneurons also exhibited more mature electrophysiological characteristics compared to controls. Overall, our studies suggest a critical role for CNS vasculature in regulating interneuron developmental maturation in both mice and humans.
Collapse
Affiliation(s)
- Matthieu Genestine
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Daisy Ambriz
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Gregg W Crabtree
- Department of Neurology, Columbia University Irving Medical CenterNew YorkUnited States
| | - Patrick Dummer
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Anna Molotkova
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Michael Quintero
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Saptarshi Biswas
- Department of Neurology, Columbia University Irving Medical CenterNew YorkUnited States
| | - Huijuan Feng
- Department of Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
| | - Chaolin Zhang
- Department of Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| | - Dritan Agalliu
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
- Department of Neurology, Columbia University Irving Medical CenterNew YorkUnited States
| | - Joseph A Gogos
- Department of Cellular Physiology and Biophysics, Columbia UniversityNew YorkUnited States
- Department of Neuroscience, Zuckerman Mind Brain and Behavior Institute, Columbia UniversityNew YorkUnited States
| | - Edmund Au
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
- Columbia Translational Neuroscience Initiative ScholarNew YorkUnited States
| |
Collapse
|
20
|
Cao KX, Ma ML, Wang CZ, Iqbal J, Si JJ, Xue YX, Yang JL. TMS-EEG: An emerging tool to study the neurophysiologic biomarkers of psychiatric disorders. Neuropharmacology 2021; 197:108574. [PMID: 33894219 DOI: 10.1016/j.neuropharm.2021.108574] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/08/2021] [Accepted: 04/15/2021] [Indexed: 01/02/2023]
Abstract
The etiology of psychiatric disorders remains largely unknown. The exploration of the neurobiological mechanisms of mental illness helps improve diagnostic efficacy and develop new therapies. This review focuses on the application of concurrent transcranial magnetic stimulation and electroencephalography (TMS-EEG) in various mental diseases, including major depressive disorder, bipolar disorder, schizophrenia, autism spectrum disorder, attention-deficit/hyperactivity disorder, substance use disorder, and insomnia. First, we summarize the commonly used protocols and output measures of TMS-EEG; then, we review the literature exploring the alterations in neural patterns, particularly cortical excitability, plasticity, and connectivity alterations, and studies that predict treatment responses and clinical states in mental disorders using TMS-EEG. Finally, we discuss the potential mechanisms underlying TMS-EEG in establishing biomarkers for psychiatric disorders and future research directions.
Collapse
Affiliation(s)
- Ke-Xin Cao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Mao-Liang Ma
- Department of Clinical Psychology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
| | - Cheng-Zhan Wang
- Department of Clinical Psychology, Tianjin Medical University General Hospital, Tianjin, China
| | - Javed Iqbal
- School of Psychology, Shaanxi Normal University and Key Laboratory for Behavior and Cognitive Neuroscience of Shaanxi Province, Xi'an, China
| | - Ji-Jian Si
- Department of Clinical Psychology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan-Xue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China; Key Laboratory for Neuroscience of Ministry of Education and Neuroscience, National Health and Family Planning Commission, Peking University, Beijing, China.
| | - Jian-Li Yang
- Department of Clinical Psychology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
21
|
Zhang L, Peng Z, Bian W, Zhu P, Tang B, Liao WP, Su T. Functional Differences Between Two Kv1.1 RNA Editing Isoforms: a Comparative Study on Neuronal Overexpression in Mouse Prefrontal Cortex. Mol Neurobiol 2021; 58:2046-2060. [PMID: 33411244 DOI: 10.1007/s12035-020-02229-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
The Shaker-related potassium channel Kv1.1 subunit has important implications for controlling neuronal excitabilities. A particular recoding by A-to-I RNA editing at I400 of Kv1.1 mRNA is an underestimated mechanism for fine-tuning the properties of Kv1.1-containing channels. Knowledge about functional differences between edited (I400V) and non-edited Kv1.1 isoforms is insufficient, especially in neurons. To understand their different roles, the two Kv1.1 isoforms were overexpressed in the prefrontal cortex via local adeno-associated virus-mediated gene delivery. The I400V isoform showed a higher competitiveness in membrane translocalization, but failed to reduce current-evoked discharges and showed weaker impact on spiking-frequency adaptation in the transduced neurons. The non-edited Kv1.1 overexpression led to slight elevations in both fast- and non-inactivating current components of macroscopic potassium current. By contrast, the I400V overexpression did not impact the fast-inactivating current component. Further isolation of Kv1.1-specific current by its specific blocker dendrotoxin-κ showed that both isoforms did result in significant increases in current amplitude, whereas the I400V was less efficient in contributing the fast-inactivating current component. Voltage-dependent properties of the fast-inactivating current component did not alter for both isoforms. For recovery kinetics, the I400V showed a significant acceleration of recovery from fast inactivation. The gene delivery of the I400V rather than the wild type exhibited anxiolytic activities, which was assessed by an open field test. These results suggest that the Kv1.1 RNA editing isoforms have different properties and outcomes, reflecting the functional and phenotypic significance of the Kv1.1 RNA editing in neurons.
Collapse
Affiliation(s)
- Liting Zhang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China.,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Zetong Peng
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China.,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Wenjun Bian
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China.,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Pingping Zhu
- Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Bin Tang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China.,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Wei-Ping Liao
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China.,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Tao Su
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China. .,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China.
| |
Collapse
|
22
|
Xu X, Song L, Hanganu-Opatz IL. Knock-Down of Hippocampal DISC1 in Immune-Challenged Mice Impairs the Prefrontal-Hippocampal Coupling and the Cognitive Performance Throughout Development. Cereb Cortex 2021; 31:1240-1258. [PMID: 33037815 PMCID: PMC7786359 DOI: 10.1093/cercor/bhaa291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/21/2020] [Accepted: 09/07/2020] [Indexed: 12/31/2022] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) gene represents an intracellular hub of developmental processes. When combined with early environmental stressors, such as maternal immune activation, but not in the absence of thereof, whole-brain DISC1 knock-down leads to memory and executive deficits as result of impaired prefrontal–hippocampal communication throughout development. While synaptic dysfunction in neonatal prefrontal cortex (PFC) has been recently identified as one source of abnormal long-range coupling, the contribution of hippocampus (HP) is still unknown. Here, we aim to fill this knowledge gap by combining in vivo electrophysiology and optogenetics with morphological and behavioral assessment of immune-challenged mice with DISC1 knock-down either in the whole brain (GE) or restricted to pyramidal neurons in hippocampal CA1 area (GHPE). We found abnormal network activity, sharp-waves, and neuronal firing in CA1 that complement the deficits in upper layer of PFC. Moreover, optogenetic activating CA1 pyramidal neurons fails to activate the prefrontal local circuits. These deficits that persist till prejuvenile age relate to dendrite sparsification and loss of spines of CA1 pyramidal neurons. As a long-term consequence, DISC1 knock-down in HP leads to poorer recognition memory at prejuvenile age. Thus, DISC1-controlled developmental processes in HP in immune-challenged mice are critical for circuit function and cognitive behavior.
Collapse
Affiliation(s)
- Xiaxia Xu
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Lingzhen Song
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
23
|
Chen F, Danladi J, Ardalan M, Nyengaard JR, Sanchez C, Wegener G. The rat hippocampal gliovascular system following one week vortioxetine and fluoxetine. Eur Neuropsychopharmacol 2021; 42:45-56. [PMID: 33199100 DOI: 10.1016/j.euroneuro.2020.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 10/03/2020] [Accepted: 11/02/2020] [Indexed: 11/25/2022]
Abstract
We have previously reported that vortioxetine, unlike the selective serotonin reuptake inhibitor fluoxetine, produces a rapid increase of dendritic spine number and Brain Derived Neurotrophic Factor (BDNF)-associated formation of synapses with mitochondrial support in the rat hippocampal CA1 and dentate gyrus. As a continuation of this line of research, and given the putative role of brain glial cells in mediating antidepressant responses the present study investigated early effects of vortioxetine on hippocampal microvasculature and Vascular Endothelial Growth Factor (VEGF) and astrocytes and microglia cells. Rats were treated for 1 week with vortioxetine (1.6 g/kg food chow) or fluoxetine (160 mg/L drinking water) at pharmacologically relevant doses. Stereological principles were used to estimate the number of ALDH1L1 positive astrocytes and Iba1 positive microglia cells, and the length of microvessels in subregions of hippocampus. VEGF protein levels were visualized with immunohistochemistry. Our results showed that vortioxetine significantly increased the number of ramified (resting) microglia and astrocytes accompanied by VEGF level elevation, whereas fluoxetine had no effect after 7 days treatment on these measures. Our findings suggest that astrocytes and microglia may have a role in mediating the pharmacological effects of vortioxetine in rats and that these effects are mediated through mechanisms that go beyond inhibition of the serotonin transporter and may target specific 5-HT receptors. It remains to be investigated whether these findings are relevant for the therapeutic effects of vortioxetine.
Collapse
Affiliation(s)
- Fenghua Chen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Nørrebrogade 44, Building 2B, 8000 Aarhus, Denmark; Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Jibrin Danladi
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Nørrebrogade 44, Building 2B, 8000 Aarhus, Denmark
| | - Maryam Ardalan
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Nørrebrogade 44, Building 2B, 8000 Aarhus, Denmark; Department of Clinical Medicine - Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Connie Sanchez
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Nørrebrogade 44, Building 2B, 8000 Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Nørrebrogade 44, Building 2B, 8000 Aarhus, Denmark; Center of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa; AUGUST Centre, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| |
Collapse
|
24
|
Galaj E, Guo C, Huang D, Ranaldi R, Ma YY. Contrasting effects of adolescent and early-adult ethanol exposure on prelimbic cortical pyramidal neurons. Drug Alcohol Depend 2020; 216:108309. [PMID: 32998090 PMCID: PMC7814343 DOI: 10.1016/j.drugalcdep.2020.108309] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND Adolescence and early-adulthood are vulnerable developmental periods during which binge drinking can have long-lasting effects on brain function. However, little is known about the effects of binge drinking on the pyramidal cells of the prelimbic cortex (PrL) during early and protracted withdrawal periods. METHODS In the present study, we performed whole-cell patch clamp recordings and dendritic spine staining to examine the intrinsic excitability, spontaneous excitatory post-synaptic currents (sEPSCs), and spine morphology of pyramidal cells in the PrL from rats exposed to chronic intermittent ethanol (CIE) during adolescence or early-adulthood. RESULTS Compared to chronic intermittent water (CIW)-treated controls, the excitability of PrL-L5 pyramidal neurons was significantly increased 21 days after adolescent CIE but decreased 21 days after early-adult CIE. No changes of excitability in PrL Layer (L) 5 were detected 2 days after either adolescent or early-adulthood CIE. Interestingly, decreases in sEPSC amplitude and increases in thin spines ratio were detected 2 days after adolescent CIE. Furthermore, decreased frequency and amplitude of sEPSCs, accompanied by a decrease in the density of total spines and non-thin spines were observed 21 days after adolescent CIE. In contrast, increased frequency and amplitude of sEPSCs, accompanied by increased densities of total spines and non-thin spines were found 21 days after early adult CIE. CONCLUSION CIE produced prolonged neuronal and synaptic alterations in PrL-L5, and the developmental stage, i.e., adolescence vs. early-adulthood when subjects receive CIE, is a key factor in determining the direction of these changes.
Collapse
Affiliation(s)
- Ewa Galaj
- Department of Psychology, Behavioral Neuroscience Program, State University of New York, Binghamton, NY, 13902, USA
| | - Changyong Guo
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Donald Huang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Robert Ranaldi
- Department of Psychology, Queens College, City University of New York, Flushing, NY, 11367, USA
| | - Yao-Ying Ma
- Department of Psychology, Behavioral Neuroscience Program, State University of New York, Binghamton, NY, 13902, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
25
|
Dysregulated Glial Differentiation in Schizophrenia May Be Relieved by Suppression of SMAD4- and REST-Dependent Signaling. Cell Rep 2020; 27:3832-3843.e6. [PMID: 31242417 DOI: 10.1016/j.celrep.2019.05.088] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 04/04/2019] [Accepted: 05/22/2019] [Indexed: 12/22/2022] Open
Abstract
Astrocytic differentiation is developmentally impaired in patients with childhood-onset schizophrenia (SCZ). To determine why, we used genetic gain- and loss-of-function studies to establish the contributions of differentially expressed transcriptional regulators to the defective differentiation of glial progenitor cells (GPCs) produced from SCZ patient-derived induced pluripotent cells (iPSCs). Negative regulators of the bone morphogenetic protein (BMP) pathway were upregulated in SCZ GPCs, including BAMBI, FST, and GREM1, whose overexpression retained SCZ GPCs at the progenitor stage. SMAD4 knockdown (KD) suppressed the production of these BMP inhibitors by SCZ GPCs and rescued normal astrocytic differentiation. In addition, the BMP-regulated transcriptional repressor REST was upregulated in SCZ GPCs, and its KD similarly restored normal glial differentiation. REST KD also rescued potassium-transport-associated gene expression and K+ uptake, which were otherwise deficient in SCZ glia. These data suggest that the glial differentiation defect in childhood-onset SCZ, and its attendant disruption in K+ homeostasis, may be rescued by targeting BMP/SMAD4- and REST-dependent transcription.
Collapse
|
26
|
Zhou C, Kong D, Xue R, Chen M, Li G, Xu Y, Liu S, Tian H, Zhuo C. Metformin Enhances Antidepressant/Antipsychotic Combination Therapy of Schizophrenia With Comorbid Depression in a Murine Model. Front Neurosci 2020; 14:517. [PMID: 32581680 PMCID: PMC7283619 DOI: 10.3389/fnins.2020.00517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 04/27/2020] [Indexed: 01/01/2023] Open
Abstract
Comorbid depressive disorders confound the diagnosis and therapy of schizophrenia. Using a murine model incorporating both MK801 and chronic unpredictable mild stress exposures, we successfully replicated both psychosis and depression. Ex vivo patch clamp recordings and in vivo calcium imaging demonstrated impaired neural activity in the prefrontal cortex (PFC). We then administered triple-drug combinations consisting of two antidepressants (mirtazapine and venlafaxine) plus an antipsychotic (either clozapine or olanzapine), and found improved PFC neuronal activity and performance in behavioral assays. Moreover, the addition of metformin to both psychotropic drug combinations brought further improvements in depressive and schizophrenic-like behaviors and physiological parameters. In summary, our data modeled the neuropathophysiology of schizophrenia with comorbid depression, and may inform drug intervention strategies.
Collapse
Affiliation(s)
- Chunhua Zhou
- Department of Pharmacology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Dezhi Kong
- Two-Photon In Vivo Imaging Centre, Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, China
| | - Rong Xue
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Min Chen
- Department of Psychiatry, School of Mental Health, Jining Medical University, Jining, China
| | - Gongying Li
- Department of Psychiatry, School of Mental Health, Jining Medical University, Jining, China
| | - Yong Xu
- MDT Center for Cognitive Impairment and Sleep Disorders, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Sha Liu
- MDT Center for Cognitive Impairment and Sleep Disorders, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Hongjun Tian
- Psychiatric-Neurological-Imaging-Laboratory, Tianjin Medical University Fourth Central Hospital, Tianjin Fourth Center Hospital, Tianjin, China
| | - Chuanjun Zhuo
- Psychiatric-Neurological-Imaging-Laboratory, Tianjin Medical University Fourth Central Hospital, Tianjin Fourth Center Hospital, Tianjin, China.,Department of Psychiatry, School of Mental Health, Jining Medical University, Jining, China.,MDT Center for Cognitive Impairment and Sleep Disorders, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
27
|
Zhang BY, Zhang YL, Sun Q, Zhang PA, Wang XX, Xu GY, Hu J, Zhang HH. Alpha-lipoic acid downregulates TRPV1 receptor via NF-κB and attenuates neuropathic pain in rats with diabetes. CNS Neurosci Ther 2020; 26:762-772. [PMID: 32175676 PMCID: PMC7298987 DOI: 10.1111/cns.13303] [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: 11/28/2019] [Revised: 02/17/2020] [Accepted: 03/01/2020] [Indexed: 12/14/2022] Open
Abstract
Aims Painful diabetic neuropathy (PDN) is a refractory complication of diabetes. The study aimed to investigate the role of α‐lipoic acid (ALA) on the regulation of transient receptor potential vanilloid‐1 (TRPV1) in dorsal root ganglion (DRG) neurons of rats with diabetes. Methods Whole‐cell patch‐clamp recordings were employed to measure neuronal excitability in DiI‐labeled DRG neurons of control and streptozotocin (STZ)‐induced diabetic rats. Western blotting and immunofluorescence assays were used to determine the expression and location of NF‐κBp65 and TRPV1. Results STZ‐induced hindpaw pain hypersensitivity and neuronal excitability in L4‐6 DRG neurons were attenuated by intraperitoneal injection with ALA once a day lasted for one week. TRPV1 expression was enhanced in L4‐6 DRGs of diabetic rats compared with age‐matched control rats, which was also suppressed by ALA treatment. In addition, TRPV1 and p65 colocated in the same DRG neurons. The expression of p65 was upregulated in L4‐6 DRGs of diabetic rats. Inhibition of p65 signaling using recombinant lentiviral vectors designated as LV‐NF‐κBp65 siRNA remarkably suppressed TRPV1 expression. Finally, p65 expression was downregulated by ALA treatment. Conclusion Our findings demonstrated that ALA may alleviate neuropathic pain in diabetes by regulating TRPV1 expression via affecting NF‐κB.
Collapse
Affiliation(s)
- Bing-Yu Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Yi-Lian Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Qian Sun
- Center for Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Ping-An Zhang
- Center for Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xi-Xi Wang
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Guang-Yin Xu
- Center for Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Hong-Hong Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| |
Collapse
|
28
|
Gogos JA, Crabtree G, Diamantopoulou A. The abiding relevance of mouse models of rare mutations to psychiatric neuroscience and therapeutics. Schizophr Res 2020; 217:37-51. [PMID: 30987923 PMCID: PMC6790166 DOI: 10.1016/j.schres.2019.03.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 01/08/2023]
Abstract
Studies using powerful family-based designs aided by large scale case-control studies, have been instrumental in cracking the genetic complexity of the disease, identifying rare and highly penetrant risk mutations and providing a handle on experimentally tractable model systems. Mouse models of rare mutations, paired with analysis of homologous cognitive and sensory processing deficits and state-of-the-art neuroscience methods to manipulate and record neuronal activity have started providing unprecedented insights into pathogenic mechanisms and building the foundation of a new biological framework for understanding mental illness. A number of important principles are emerging, namely that degradation of the computational mechanisms underlying the ordered activity and plasticity of both local and long-range neuronal assemblies, the building blocks necessary for stable cognition and perception, might be the inevitable consequence and the common point of convergence of the vastly heterogeneous genetic liability, manifesting as defective internally- or stimulus-driven neuronal activation patterns and triggering the constellation of schizophrenia symptoms. Animal models of rare mutations have the unique potential to help us move from "which" (gene) to "how", "where" and "when" computational regimes of neural ensembles are affected. Linking these variables should improve our understanding of how symptoms emerge and how diagnostic boundaries are established at a circuit level. Eventually, a better understanding of pathophysiological trajectories at the level of neural circuitry in mice, aided by basic human experimental biology, should guide the development of new therapeutics targeting either altered circuitry itself or the underlying biological pathways.
Collapse
Affiliation(s)
- Joseph A. Gogos
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027 USA,Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA,Department of Neuroscience, Columbia University, New York, NY 10032 USA,Correspondence should be addressed to: Joseph A. Gogos ()
| | - Gregg Crabtree
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027 USA,Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Anastasia Diamantopoulou
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027 USA,Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
29
|
Cognition- and circuit-based dysfunction in a mouse model of 22q11.2 microdeletion syndrome: effects of stress. Transl Psychiatry 2020; 10:41. [PMID: 32066701 PMCID: PMC7026063 DOI: 10.1038/s41398-020-0687-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 11/19/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022] Open
Abstract
Genetic microdeletion at the 22q11 locus is associated with very high risk for schizophrenia. The 22q11.2 microdeletion (Df(h22q11)/+) mouse model shows cognitive deficits observed in this disorder, some of which can be linked to dysfunction of the prefrontal cortex (PFC). We used behavioral (n = 10 per genotype), electrophysiological (n = 7 per genotype per group), and neuroanatomical (n = 5 per genotype) techniques to investigate schizophrenia-related pathology of Df(h22q11)/+ mice, which showed a significant decrease in the total number of parvalbumin positive interneurons in the medial PFC. The Df(h22q11)/+ mice when tested on PFC-dependent behavioral tasks, including gambling tasks, perform significantly worse than control animals while exhibiting normal behavior on hippocampus-dependent tasks. They also show a significant decrease in hippocampus-medial Prefrontal cortex (H-PFC) synaptic plasticity (long-term potentiation, LTP). Acute platform stress almost abolished H-PFC LTP in both wild-type and Df(h22q11)/+ mice. H-PFC LTP was restored to prestress levels by clozapine (3 mg/kg i.p.) in stressed Df(h22q11)/+ mice, but the restoration of stress-induced LTP, while significant, was similar between wild-type and Df(h22q11)/+ mice. A medial PFC dysfunction may underlie the negative and cognitive symptoms in human 22q11 deletion carriers, and these results are relevant to the current debate on the utility of clozapine in such subjects.
Collapse
|
30
|
Mi Z, Yang J, He Q, Zhang X, Xiao Y, Shu Y. Alterations of Electrophysiological Properties and Ion Channel Expression in Prefrontal Cortex of a Mouse Model of Schizophrenia. Front Cell Neurosci 2019; 13:554. [PMID: 31920555 PMCID: PMC6927988 DOI: 10.3389/fncel.2019.00554] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/02/2019] [Indexed: 11/13/2022] Open
Abstract
Maternal immune activation (MIA) and juvenile social isolation (SI) are two most prevalent and widely accepted environmental insults that could increase the propensity of psychiatric illnesses. Using a two-hit mouse model, we examined the impact of the combination of these two factors on animal behaviors, neuronal excitability and expressions of voltage-gated sodium (Nav) and small conductance calcium-activated potassium (SK) channels in the prefrontal cortex (PFC). We found that MIA-SI induced a number of schizophrenia-related behavioral deficits. Patch clamp recordings revealed alterations in electrophysiological properties of PFC layer-5 pyramidal cells, including hyperpolarized resting membrane potential (RMP), increased input resistance and enhanced medium after-hyperpolarization (mAHP). MIA-SI also increased the ratio of the maximal slope of somatodendritic potential to the peak slope of action potential upstroke, indicating a change in perisomatic Nav availability. Consistently, MIA-SI significantly increased the expression level of Nav1.2 and SK3 channels that contribute to the somatodendritic potential and the mAHP, respectively. Together, these changes may alter neuronal signaling in the PFC and behavioral states, representing a molecular imprint of environmental insults associated with neuropsychiatric illnesses.
Collapse
Affiliation(s)
- Zhen Mi
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Jun Yang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Quansheng He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Xiaowen Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Yujie Xiao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Yousheng Shu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| |
Collapse
|
31
|
Klorig DC, Alberto GE, Smith T, Godwin DW. Optogenetically-Induced Population Discharge Threshold as a Sensitive Measure of Network Excitability. eNeuro 2019; 6:ENEURO.0229-18.2019. [PMID: 31619450 PMCID: PMC6838688 DOI: 10.1523/eneuro.0229-18.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/06/2019] [Accepted: 09/27/2019] [Indexed: 12/21/2022] Open
Abstract
Network excitability is governed by synaptic efficacy, intrinsic excitability, and the circuitry in which these factors are expressed. The complex interplay between these factors determines how circuits function and, at the extreme, their susceptibility to seizure. We have developed a sensitive, quantitative estimate of network excitability in freely behaving mice using a novel optogenetic intensity-response procedure. Synchronous activation of deep sublayer CA1 pyramidal cells produces abnormal network-wide epileptiform population discharges (PDs) that are nearly indistinguishable from spontaneously-occurring interictal spikes (IISs). By systematically varying light intensity, and therefore the magnitude of the optogenetically-mediated current, we generated intensity-response curves using the probability of PD as the dependent variable. Manipulations known to increase excitability, such as sub-convulsive doses (20 mg/kg) of the chemoconvulsant pentylenetetrazol (PTZ), produced a leftward shift in the curve compared to baseline. The anti-epileptic drug levetiracetam (LEV; 40 mk/kg), in combination with PTZ, produced a rightward shift. Optogenetically-induced PD threshold (oPDT) baselines were stable over time, suggesting the metric is appropriate for within-subject experimental designs with multiple pharmacological manipulations.
Collapse
Affiliation(s)
- D C Klorig
- Department of Neurobiology and Anatomy
- Neuroscience Program
| | - G E Alberto
- Department of Neurobiology and Anatomy
- Neuroscience Program
| | - T Smith
- Department of Neurobiology and Anatomy
| | - D W Godwin
- Department of Neurobiology and Anatomy
- Neuroscience Program
- Department of Physiology and Pharmacology, Wake Forest University, Winston-Salem, NC
| |
Collapse
|
32
|
Gastrin-releasing peptide inhibits CA1 neurons via increasing inhibitory synaptic transmissions in hippocampal slices of rats. Neuroreport 2019; 30:1048-1053. [PMID: 31490840 DOI: 10.1097/wnr.0000000000001324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Gastrin-releasing peptide plays an important role in regulating the advanced functions of the brain including emotional behavior, learning and memory. What's more, gastrin-releasing peptide levels are also associated with the central nervous system diseases. Our previous study proposed that intraperitoneal injection of gastrin-releasing peptide can improve spatial memory in chronic ischemic model rats. It is well known that the hippocampus is an important brain area related to spatial learning and memory, but the mechanisms of gastrin-releasing peptide on hippocampal neurons are still unclear. In this study, we examined the effects of gastrin-releasing peptide on excitability of hippocampal CA1 neurons and further explored the mechanisms of its effects on synaptic transmission. The results showed that gastrin-releasing peptide inhibited the excitability of CA1 neurons and increased the amplitude and frequency of inhibitory postsynaptic currents significantly. In summary, we demonstrate that gastrin-releasing peptide can inhibit the excitability of hippocampal CA1 area neurons in brain slices and clarify the synaptic transmission mechanism involved in this process, which provide a theoretical basis for gastrin-releasing peptide to improve animal cognitive function, and new ideas for the treatment of related central nervous system diseases.
Collapse
|
33
|
Mukai J, Cannavò E, Crabtree GW, Sun Z, Diamantopoulou A, Thakur P, Chang CY, Cai Y, Lomvardas S, Takata A, Xu B, Gogos JA. Recapitulation and Reversal of Schizophrenia-Related Phenotypes in Setd1a-Deficient Mice. Neuron 2019; 104:471-487.e12. [PMID: 31606247 DOI: 10.1016/j.neuron.2019.09.014] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 07/28/2019] [Accepted: 09/10/2019] [Indexed: 12/15/2022]
Abstract
SETD1A, a lysine-methyltransferase, is a key schizophrenia susceptibility gene. Mice carrying a heterozygous loss-of-function mutation of the orthologous gene exhibit alterations in axonal branching and cortical synaptic dynamics accompanied by working memory deficits. We show that Setd1a binds both promoters and enhancers with a striking overlap between Setd1a and Mef2 on enhancers. Setd1a targets are highly expressed in pyramidal neurons and display a complex pattern of transcriptional up- and downregulations shaped by presumed opposing functions of Setd1a on promoters and Mef2-bound enhancers. Notably, evolutionarily conserved Setd1a targets are associated with neuropsychiatric genetic risk burden. Reinstating Setd1a expression in adulthood rescues cognitive deficits. Finally, we identify LSD1 as a major counteracting demethylase for Setd1a and show that its pharmacological antagonism results in a full rescue of the behavioral and morphological deficits in Setd1a-deficient mice. Our findings advance understanding of how SETD1A mutations predispose to schizophrenia (SCZ) and point to novel therapeutic interventions.
Collapse
Affiliation(s)
- Jun Mukai
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Enrico Cannavò
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Gregg W Crabtree
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Ziyi Sun
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Anastasia Diamantopoulou
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Pratibha Thakur
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA
| | - Chia-Yuan Chang
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yifei Cai
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Stavros Lomvardas
- Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Neuroscience, Columbia University, New York, NY 10032, USA
| | - Atsushi Takata
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Bin Xu
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - Joseph A Gogos
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute Columbia University, New York, NY 10027, USA; Department of Neuroscience, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
34
|
Mäki-Marttunen T, Devor A, Phillips WA, Dale AM, Andreassen OA, Einevoll GT. Computational Modeling of Genetic Contributions to Excitability and Neural Coding in Layer V Pyramidal Cells: Applications to Schizophrenia Pathology. Front Comput Neurosci 2019; 13:66. [PMID: 31616272 PMCID: PMC6775251 DOI: 10.3389/fncom.2019.00066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 09/09/2019] [Indexed: 11/13/2022] Open
Abstract
Pyramidal cells in layer V of the neocortex are one of the most widely studied neuron types in the mammalian brain. Due to their role as integrators of feedforward and cortical feedback inputs, they are well-positioned to contribute to the symptoms and pathology in mental disorders-such as schizophrenia-that are characterized by a mismatch between the internal perception and external inputs. In this modeling study, we analyze the input/output properties of layer V pyramidal cells and their sensitivity to modeled genetic variants in schizophrenia-associated genes. We show that the excitability of layer V pyramidal cells and the way they integrate inputs in space and time are altered by many types of variants in ion-channel and Ca2+ transporter-encoding genes that have been identified as risk genes by recent genome-wide association studies. We also show that the variability in the output patterns of spiking and Ca2+ transients in layer V pyramidal cells is altered by these model variants. Importantly, we show that many of the predicted effects are robust to noise and qualitatively similar across different computational models of layer V pyramidal cells. Our modeling framework reveals several aspects of single-neuron excitability that can be linked to known schizophrenia-related phenotypes and existing hypotheses on disease mechanisms. In particular, our models predict that single-cell steady-state firing rate is positively correlated with the coding capacity of the neuron and negatively correlated with the amplitude of a prepulse-mediated adaptation and sensitivity to coincidence of stimuli in the apical dendrite and the perisomatic region of a layer V pyramidal cell. These results help to uncover the voltage-gated ion-channel and Ca2+ transporter-associated genetic underpinnings of schizophrenia phenotypes and biomarkers.
Collapse
Affiliation(s)
| | - Anna Devor
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States.,Department of Radiology, University of California San Diego, La Jolla, CA, United States.,Martinos Center for Biomedical Imaging, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - William A Phillips
- Psychology, Faculty of Natural Sciences, University of Stirling, Stirling, United Kingdom
| | - Anders M Dale
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States.,Department of Radiology, University of California San Diego, La Jolla, CA, United States
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gaute T Einevoll
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway.,Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
35
|
Bartley AF, Abiraman K, Stewart LT, Hossain MI, Gahan DM, Kamath AV, Burdette MK, Andrabe S, Foulger SH, McMahon LL, Dobrunz LE. LSO:Ce Inorganic Scintillators Are Biocompatible With Neuronal and Circuit Function. Front Synaptic Neurosci 2019; 11:24. [PMID: 31551750 PMCID: PMC6733890 DOI: 10.3389/fnsyn.2019.00024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Optogenetics is widely used in neuroscience to control neural circuits. However, non-invasive methods for light delivery in brain are needed to avoid physical damage caused by current methods. One potential strategy could employ x-ray activation of radioluminescent particles (RPLs), enabling localized light generation within the brain. RPLs composed of inorganic scintillators can emit light at various wavelengths depending upon composition. Cerium doped lutetium oxyorthosilicate (LSO:Ce), an inorganic scintillator that emits blue light in response to x-ray or ultraviolet (UV) stimulation, could potentially be used to control neural circuits through activation of channelrhodopsin-2 (ChR2), a light-gated cation channel. Whether inorganic scintillators themselves negatively impact neuronal processes and synaptic function is unknown, and was investigated here using cellular, molecular, and electrophysiological approaches. As proof of principle, we applied UV stimulation to 4 μm LSO:Ce particles during whole-cell recording of CA1 pyramidal cells in acute hippocampal slices from mice that expressed ChR2 in glutamatergic neurons. We observed an increase in frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs), indicating activation of ChR2 and excitation of neurons. Importantly, LSO:Ce particles did not affect survival of primary mouse cortical neurons, even after 24 h of exposure. In extracellular dendritic field potential recordings, no change in the strength of basal glutamatergic transmission was observed during exposure to LSO:Ce microparticles. However, the amplitude of the fiber volley was slightly reduced with high stimulation. Additionally, there was a slight decrease in the frequency of sEPSCs in whole-cell voltage-clamp recordings from CA1 pyramidal cells, with no change in current amplitudes. The amplitude and frequency of spontaneous inhibitory postsynaptic currents were unchanged. Finally, long term potentiation (LTP), a synaptic modification believed to underlie learning and memory and a robust measure of synaptic integrity, was successfully induced, although the magnitude was slightly reduced. Together, these results show LSO:Ce particles are biocompatible even though there are modest effects on baseline synaptic function and long-term synaptic plasticity. Importantly, we show that light emitted from LSO:Ce particles is able to activate ChR2 and modify synaptic function. Therefore, LSO:Ce inorganic scintillators are potentially viable for use as a new light delivery system for optogenetics.
Collapse
Affiliation(s)
- Aundrea F. Bartley
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kavitha Abiraman
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Luke T. Stewart
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mohammed Iqbal Hossain
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - David M. Gahan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Abhishek V. Kamath
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary K. Burdette
- Department of Materials Science and Engineering, Clemson University, Anderson, SC, United States
| | - Shaida Andrabe
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stephen H. Foulger
- Department of Materials Science and Engineering, Clemson University, Anderson, SC, United States
- Center for Optical Materials Science and Engineering Technologies, Clemson University, Anderson, SC, United States
- Department of Bioengineering, Clemson University, Clemson, SC, United States
| | - Lori L. McMahon
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lynn E. Dobrunz
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Neuroscience Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
36
|
Yang K, Kondo MA, Jaaro-Peled H, Cash-Padgett T, Kano SI, Ishizuka K, Pevsner J, Tomoda T, Sawa A, Niwa M. The transcriptome landscape associated with Disrupted-in-Schizophrenia-1 locus impairment in early development and adulthood. Schizophr Res 2019; 210:149-156. [PMID: 31204062 PMCID: PMC8050833 DOI: 10.1016/j.schres.2019.05.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 01/08/2023]
Abstract
DISC1 was originally expected to be a genetic risk factor for schizophrenia, but the genome wide association studies have not supported this idea. In contrast, neurobiological studies of DISC1 in cell and animal models have demonstrated that direct perturbation of DISC1 protein elicits neurobiological and behavioral abnormalities relevant to a wide range of psychiatric conditions, in particular psychosis. Thus, the utility of DISC1 as a biological lead for psychosis research is clear. In the present study, we aimed to capture changes in the molecular landscape in the prefrontal cortex upon perturbation of DISC1, using the Disc1 locus impairment (Disc1-LI) model in which the majority of Disc1 isoforms have been depleted, and to explore potential molecular mediators relevant to psychiatric conditions. We observed a robust change in gene expression profile elicited by Disc1-LI in which the stronger effects on molecular networks were observed in early stage compared with those in adulthood. Significant alterations were found in specific pathways relevant to psychiatric conditions, such as pathways of signaling by G protein-coupled receptor, neurotransmitter release cycle, and voltage gated potassium channels. The differentially expressed genes (DEGs) between Disc1-LI and wild-type mice are significantly enriched not only in neurons, but also in astrocytes and oligodendrocyte precursor cells. The brain-disorder-associated genes at the mRNA and protein levels rather than those at the genomic levels are enriched in the DEGs. Together, our present study supports the utility of Disc1-LI mice in biological research for psychiatric disorder-associated molecular networks.
Collapse
Affiliation(s)
- Kun Yang
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mari A Kondo
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hanna Jaaro-Peled
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tyler Cash-Padgett
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Shin-Ichi Kano
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Koko Ishizuka
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jonathan Pevsner
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Toshifumi Tomoda
- Medical Innovation Center, Kyoto University, Kyoto 606-8397, Japan
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Mental Health, Johns Hopkins University Bloomberg School of Medicine, Baltimore, MD 21205, USA.
| | - Minae Niwa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
37
|
Oberlander VC, Xu X, Chini M, Hanganu-Opatz IL. Developmental dysfunction of prefrontal-hippocampal networks in mouse models of mental illness. Eur J Neurosci 2019; 50:3072-3084. [PMID: 31087437 PMCID: PMC6851774 DOI: 10.1111/ejn.14436] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/18/2019] [Accepted: 05/08/2019] [Indexed: 12/28/2022]
Abstract
Despite inherent difficulties to translate human cognitive phenotype into animals, a large number of animal models for psychiatric disorders, such as schizophrenia, have been developed over the last decades. To which extent they reproduce common patterns of dysfunction related to mental illness and abnormal processes of maturation is still largely unknown. While the devastating symptoms of disease are firstly detectable in adulthood, they are considered to reflect profound miswiring of brain circuitry as result of abnormal development. To reveal whether different disease models share common dysfunction early in life, we investigate the prefrontal-hippocampal communication at neonatal age in (a) mice mimicking the abnormal genetic background (22q11.2 microdeletion, DISC1 knockdown), (b) mice mimicking the challenge by environmental stressors (maternal immune activation during pregnancy), (c) mice mimicking the combination of both aetiologies (dual-hit models) and pharmacological mouse models. Simultaneous extracellular recordings in vivo from all layers of prelimbic subdivision (PL) of prefrontal cortex (PFC) and CA1 area of intermediate/ventral hippocampus (i/vHP) show that network oscillations have a more fragmented structure and decreased power mainly in neonatal mice that mimic both genetic and environmental aetiology of disease. These mice also show layer-specific firing deficits in PL. Similar early network dysfunction was present in mice with 22q11.2 microdeletion. The abnormal activity patterns are accompanied by weaker synchrony and directed interactions within prefrontal-hippocampal networks. Thus, only severe genetic defects or combined genetic environmental stressors are disruptive enough for reproducing the early network miswiring in mental disorders.
Collapse
Affiliation(s)
- Victoria C Oberlander
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Xiaxia Xu
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mattia Chini
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Developmental Neurophysiology, Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
38
|
Johnstone M, Hillary RF, St Clair D. Stem Cells to Inform the Neurobiology of Mental Illness. Curr Top Behav Neurosci 2019; 40:13-43. [PMID: 30030769 DOI: 10.1007/7854_2018_57] [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] [Indexed: 12/13/2022]
Abstract
The inception of human-induced pluripotent stem cell (hiPSCs) technology has provided an exciting platform upon which the modelling and treatment of human neurodevelopmental and neuropsychiatric disorders may be expedited. Although the genetic architecture of these disorders is far more complex than previously imagined, many key loci have at last been identified. This has allowed in vivo and in vitro technologies to be refined to model specific high-penetrant genetic loci involved in both disorders. Animal models of neurodevelopmental disorders, such as schizophrenia and autism spectrum disorders, show limitations in recapitulating the full complexity and heterogeneity of human neurodevelopmental disease states. Indeed, patient-derived hiPSCs offer distinct advantages over classical animal models in the study of human neuropathologies. Here we have discussed the current, relative translational merit of hiPSCs in investigating human neurodevelopmental and neuropsychiatric disorders with a specific emphasis on the utility of such systems to aid in the identification of biomarkers. We have highlighted the promises and pitfalls of reprogramming cell fate for the study of these disorders and provide recommendations for future directions in this field in order to overcome current limitations. Ultimately, this will aid in the development of effective clinical strategies for diverse patient populations affected by these disorders with the aim of also leading to biomarker identification.
Collapse
Affiliation(s)
- Mandy Johnstone
- Division of Psychiatry, Royal Edinburgh Hospital, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK.
| | - Robert F Hillary
- Division of Psychiatry, Royal Edinburgh Hospital, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - David St Clair
- Division of Psychiatry, Royal Edinburgh Hospital, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
39
|
Vo BN, Abney KK, Anderson A, Marron Fernandez de Velasco E, Benneyworth MA, Daniels JS, Morrison RD, Hopkins CR, Weaver CD, Wickman K. VU0810464, a non-urea G protein-gated inwardly rectifying K + (K ir 3/GIRK) channel activator, exhibits enhanced selectivity for neuronal K ir 3 channels and reduces stress-induced hyperthermia in mice. Br J Pharmacol 2019; 176:2238-2249. [PMID: 30924523 DOI: 10.1111/bph.14671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/16/2019] [Accepted: 02/20/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE G protein-gated inwardly rectifying K+ (Kir 3) channels moderate the activity of excitable cells and have been implicated in neurological disorders and cardiac arrhythmias. Most neuronal Kir 3 channels consist of Kir 3.1 and Kir 3.2 subtypes, while cardiac Kir 3 channels consist of Kir 3.1 and Kir 3.4 subtypes. Previously, we identified a family of urea-containing Kir 3 channel activators, but these molecules exhibit suboptimal pharmacokinetic properties and modest selectivity for Kir 3.1/3.2 relative to Kir 3.1/3.4 channels. Here, we characterize a non-urea activator, VU0810464, which displays nanomolar potency as a Kir 3.1/3.2 activator, improved selectivity for neuronal Kir 3 channels, and improved brain penetration. EXPERIMENTAL APPROACH We used whole-cell electrophysiology to measure the efficacy and potency of VU0810464 in neurons and the selectivity of VU0810464 for neuronal and cardiac Kir 3 channel subtypes. We tested VU0810464 in vivo in stress-induced hyperthermia and elevated plus maze paradigms. Parallel studies with ML297, the prototypical activator of Kir 3.1-containing Kir 3 channels, were performed to permit direct comparisons. KEY RESULTS VU0810464 and ML297 exhibited comparable efficacy and potency as neuronal Kir 3 channel activators, but VU0810464 was more selective for neuronal Kir 3 channels. VU0810464, like ML297, reduced stress-induced hyperthermia in a Kir 3-dependent manner in mice. ML297, but not VU0810464, decreased anxiety-related behaviour as assessed with the elevated plus maze test. CONCLUSION AND IMPLICATIONS VU0810464 represents a new class of Kir 3 channel activator with enhanced selectivity for Kir 3.1/3.2 channels. VU0810464 may be useful for examining Kir 3.1/3.2 channel contributions to complex behaviours and for probing the potential of Kir 3 channel-dependent manipulations to treat neurological disorders.
Collapse
Affiliation(s)
- Baovi N Vo
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Kristopher K Abney
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN
| | - Allison Anderson
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | | | | | | | - Ryan D Morrison
- Research and Development, Precera Bioscience, Inc., Franklin, TN
| | - Corey R Hopkins
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
40
|
Zbili M, Debanne D. Past and Future of Analog-Digital Modulation of Synaptic Transmission. Front Cell Neurosci 2019; 13:160. [PMID: 31105529 PMCID: PMC6492051 DOI: 10.3389/fncel.2019.00160] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/08/2019] [Indexed: 01/12/2023] Open
Abstract
Action potentials (APs) are generally produced in response to complex summation of excitatory and inhibitory synaptic inputs. While it is usually considered as a digital event, both the amplitude and width of the AP are significantly impacted by the context of its emission. In particular, the analog variations in subthreshold membrane potential determine the spike waveform and subsequently affect synaptic strength, leading to the so-called analog-digital modulation of synaptic transmission. We review here the numerous evidence suggesting context-dependent modulation of spike waveform, the discovery analog-digital modulation of synaptic transmission in invertebrates and its recent validation in mammals. We discuss the potential roles of analog-digital transmission in the physiology of neural networks.
Collapse
Affiliation(s)
- Mickael Zbili
- UNIS, UMR 1072, INSERM AMU, Marseille, France.,CRNL, INSERM U1028-CNRS UMR5292-Université Claude Bernard Lyon1, Lyon, France
| | | |
Collapse
|
41
|
Masuda F, Nakajima S, Miyazaki T, Tarumi R, Ogyu K, Wada M, Tsugawa S, Croarkin PE, Mimura M, Noda Y. Clinical effectiveness of repetitive transcranial magnetic stimulation treatment in children and adolescents with neurodevelopmental disorders: A systematic review. AUTISM : THE INTERNATIONAL JOURNAL OF RESEARCH AND PRACTICE 2019; 23:1614-1629. [PMID: 30663323 DOI: 10.1177/1362361318822502] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neurodevelopmental disorders, including autism spectrum disorder, are common in children and adolescents, but treatment strategies remain limited. Although repetitive transcranial magnetic stimulation has been studied for neurodevelopmental disorders, there is no clear consensus on its therapeutic effects. This systematic review examined literature on repetitive transcranial magnetic stimulation for children and adolescents with neurodevelopmental disorders published up to 2018 using the PubMed database. The search identified 264 articles and 14 articles met eligibility criteria. Twelve of these studies used conventional repetitive transcranial magnetic stimulation and two studies used theta burst stimulation. No severe adverse effects were reported in these studies. In patients with autism spectrum disorder, low-frequency repetitive transcranial magnetic stimulation and intermittent theta burst stimulation applied to the dorsolateral prefrontal cortex may have therapeutic effects on social functioning and repetitive behaviors. In patients with attention deficit/hyperactivity disorder, low-frequency repetitive transcranial magnetic stimulation applied to the left dorsolateral prefrontal cortex and high-frequency repetitive transcranial magnetic stimulation applied to the right dorsolateral prefrontal cortex may target inattention, hyperactivity, and impulsivity. In patients with tic disorders, low-frequency repetitive transcranial magnetic stimulation applied to the bilateral supplementary motor area improved tic symptom severity. This systematic review suggests that repetitive transcranial magnetic stimulation may be a promising intervention for children and adolescents with neurodevelopmental disorders. The results warrant further large randomized controlled trials of repetitive transcranial magnetic stimulation in children with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Fumi Masuda
- 1 Keio University School of Medicine, Japan.,2 Shiga University of Medical Science, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Diamantopoulou A, Gogos JA. Neurocognitive and Perceptual Processing in Genetic Mouse Models of Schizophrenia: Emerging Lessons. Neuroscientist 2019; 25:597-619. [PMID: 30654694 DOI: 10.1177/1073858418819435] [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] [Indexed: 12/21/2022]
Abstract
During the past two decades, the number of animal models of psychiatric disorders has grown exponentially. Of these, genetic animal models that are modeled after rare but highly penetrant mutations hold great promise for deciphering critical molecular, synaptic, and neurocircuitry deficits of major psychiatric disorders, such as schizophrenia. Animal models should aim to focus on core aspects rather than capture the entire human disease. In this context, animal models with strong etiological validity, where behavioral and neurophysiological phenotypes and the features of the disease being modeled are in unambiguous homology, are being used to dissect both elementary and complex cognitive and perceptual processing deficits present in psychiatric disorders at the level of neurocircuitry, shedding new light on critical disease mechanisms. Recent progress in neuroscience along with large-scale initiatives that propose a consistent approach in characterizing these deficits across different laboratories will further enhance the efficacy of these studies that will ultimately lead to identifying new biological targets for drug development.
Collapse
Affiliation(s)
- Anastasia Diamantopoulou
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, USA.,Zuckerman Mind Brain Behavior Institute, New York, NY, USA
| | - Joseph A Gogos
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, USA.,Zuckerman Mind Brain Behavior Institute, New York, NY, USA.,Department of Neuroscience, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
43
|
Transient Knock-Down of Prefrontal DISC1 in Immune-Challenged Mice Causes Abnormal Long-Range Coupling and Cognitive Dysfunction throughout Development. J Neurosci 2019; 39:1222-1235. [PMID: 30617212 PMCID: PMC6381232 DOI: 10.1523/jneurosci.2170-18.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 02/07/2023] Open
Abstract
Compromised brain development has been hypothesized to account for mental illness. This concept was underpinned by the function of the molecule disrupted-in-schizophrenia 1 (DISC1), which represents an intracellular hub of developmental processes and has been related to cognitive dysfunction in psychiatric disorders. Mice with whole-brain DISC1 knock-down show impaired prefrontal–hippocampal function and cognitive abilities throughout development and at adulthood, especially when combined with early environmental stressors, such as maternal immune activation (MIA). However, the contribution of abnormal DISC1-driven maturation of either prefrontal cortex (PFC) or hippocampus (HP) to these deficits is still unknown. Here, we use in utero electroporation to restrict the DISC1 knock-down to prefrontal layer II/III pyramidal neurons during perinatal development and expose these mice to MIA as an environmental stressor (dual-hit GPFCE mice, both sexes). Combining in vivo electrophysiology and neuroanatomy with behavioral testing, we show that GPFCE mice at neonatal age have abnormal patterns of oscillatory activity and firing in PFC, but not HP. Abnormal firing rates in PFC of GPFCE mice relate to sparser dendritic arborization and lower spine density. Moreover, the long-range coupling within prefrontal–hippocampal networks is decreased at this age. The transient prefrontal DISC1 knock-down was sufficient to permanently perturb the prefrontal–hippocampal communication and caused poorer recognition memory performance at pre-juvenile age. Thus, developmental dysfunction of prefrontal circuitry causes long-lasting disturbances related to mental illness. SIGNIFICANCE STATEMENT Hypofrontality is considered a main cause of cognitive deficits in mental disorders, yet the underlying mechanisms are still largely unknown. During development, long before the emergence of disease symptoms, the functional coupling within the prefrontal–hippocampal network, which is the core brain circuit involved in cognitive processing, is reduced. To assess to which extent impaired prefrontal development contributes to the early dysfunction, immune-challenged mice with transient DISC1 knock-down confined to PFC were investigated in their prefrontal–hippocampal communication throughout development by in vivo electrophysiology and behavioral testing. We show that perturbing developmental processes of prefrontal layer II/III pyramidal neurons is sufficient to diminish prefrontal–hippocampal coupling and decrease the cognitive performance throughout development.
Collapse
|
44
|
St Clair D, Johnstone M. Using mouse transgenic and human stem cell technologies to model genetic mutations associated with schizophrenia and autism. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0037. [PMID: 29352035 PMCID: PMC5790834 DOI: 10.1098/rstb.2017.0037] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2017] [Indexed: 12/22/2022] Open
Abstract
Solid progress has occurred over the last decade in our understanding of the molecular genetic basis of neurodevelopmental disorders, and of schizophrenia and autism in particular. Although the genetic architecture of both disorders is far more complex than previously imagined, many key loci have at last been identified. This has allowed in vivo and in vitro technologies to be refined to model specific high-penetrant genetic loci involved in both disorders. Using the DISC1/NDE1 and CYFIP1/EIF4E loci as exemplars, we explore the opportunities and challenges of using animal models and human-induced pluripotent stem cell technologies to further understand/treat and potentially reverse the worst consequences of these debilitating disorders. This article is part of a discussion meeting issue ‘Of mice and mental health: facilitating dialogue between basic and clinical neuroscientists’.
Collapse
Affiliation(s)
- David St Clair
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Mandy Johnstone
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK.,Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
45
|
Shen X, Yeung HT, Lai KO. Application of Human-Induced Pluripotent Stem Cells (hiPSCs) to Study Synaptopathy of Neurodevelopmental Disorders. Dev Neurobiol 2018; 79:20-35. [PMID: 30304570 DOI: 10.1002/dneu.22644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/27/2018] [Accepted: 10/04/2018] [Indexed: 12/15/2022]
Abstract
Synapses are the basic structural and functional units for information processing and storage in the brain. Their diverse properties and functions ultimately underlie the complexity of human behavior. Proper development and maintenance of synapses are essential for normal functioning of the nervous system. Disruption in synaptogenesis and the consequent alteration in synaptic function have been strongly implicated to cause neurodevelopmental disorders such as autism spectrum disorders (ASDs) and schizophrenia (SCZ). The introduction of human-induced pluripotent stem cells (hiPSCs) provides a new path to elucidate disease mechanisms and potential therapies. In this review, we will discuss the advantages and limitations of using hiPSC-derived neurons to study synaptic disorders. Many mutations in genes encoding for proteins that regulate synaptogenesis have been identified in patients with ASDs and SCZ. We use Methyl-CpG binding protein 2 (MECP2), SH3 and multiple ankyrin repeat domains 3 (SHANK3) and Disrupted in schizophrenia 1 (DISC1) as examples to illustrate the promise of using hiPSCs as cellular models to elucidate the mechanisms underlying disease-related synaptopathy.
Collapse
Affiliation(s)
- Xuting Shen
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Hoi Ting Yeung
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Kwok-On Lai
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| |
Collapse
|
46
|
Tropea D, Hardingham N, Millar K, Fox K. Mechanisms underlying the role of DISC1 in synaptic plasticity. J Physiol 2018; 596:2747-2771. [PMID: 30008190 PMCID: PMC6046077 DOI: 10.1113/jp274330] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/02/2018] [Indexed: 12/11/2022] Open
Abstract
Disrupted in schizophrenia 1 (DISC1) is an important hub protein, forming multimeric complexes by self-association and interacting with a large number of synaptic and cytoskeletal molecules. The synaptic location of DISC1 in the adult brain suggests a role in synaptic plasticity, and indeed, a number of studies have discovered synaptic plasticity impairments in a variety of different DISC1 mutants. This review explores the possibility that DISC1 is an important molecule for organizing proteins involved in synaptic plasticity and examines why mutations in DISC1 impair plasticity. It concentrates on DISC1's role in interacting with synaptic proteins, controlling dendritic structure and cellular trafficking of mRNA, synaptic vesicles and mitochondria. N-terminal directed mutations appear to impair synaptic plasticity through interactions with phosphodiesterase 4B (PDE4B) and hence protein kinase A (PKA)/GluA1 and PKA/cAMP response element-binding protein (CREB) signalling pathways, and affect spine structure through interactions with kalirin 7 (Kal-7) and Rac1. C-terminal directed mutations also impair plasticity possibly through altered interactions with lissencephaly protein 1 (LIS1) and nuclear distribution protein nudE-like 1 (NDEL1), thereby affecting developmental processes such as dendritic structure and spine maturation. Many of the same molecules involved in DISC1's cytoskeletal interactions are also involved in intracellular trafficking, raising the possibility that impairments in intracellular trafficking affect cytoskeletal development and vice versa. While the multiplicity of DISC1 protein interactions makes it difficult to pinpoint a single causal signalling pathway, we suggest that the immediate-term effects of N-terminal influences on GluA1, Rac1 and CREB, coupled with the developmental effects of C-terminal influences on trafficking and the cytoskeleton make up the two main branches of DISC1's effect on synaptic plasticity and dendritic spine stability.
Collapse
Affiliation(s)
- Daniela Tropea
- Neurospychiatric GeneticsTrinity Center for Health Sciences and Trinity College Institute of Neuroscience (TCIN)Trinity College DublinDublinIreland
| | - Neil Hardingham
- School of BiosciencesMuseum AvenueCardiff UniversityCardiffUK
| | - Kirsty Millar
- Centre for Genomic & Experimental MedicineMRC Institute of Genetics & Molecular MedicineWestern General HospitalUniversity of EdinburghCrewe RoadEdinburghUK
| | - Kevin Fox
- School of BiosciencesMuseum AvenueCardiff UniversityCardiffUK
| |
Collapse
|
47
|
Cardarelli RA, Martin R, Jaaro-Peled H, Sawa A, Powell EM, O'Donnell P. Dominant-Negative DISC1 Alters the Dopaminergic Modulation of Inhibitory Interneurons in the Mouse Prefrontal Cortex. MOLECULAR NEUROPSYCHIATRY 2018; 4:20-29. [PMID: 29998115 DOI: 10.1159/000488030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 02/26/2018] [Indexed: 11/19/2022]
Abstract
A truncated disrupted in schizophrenia 1 (Disc1) gene increases the risk of psychiatric disorders, probably affecting cortical interneurons. Here, we sought to determine whether this cell population is affected in mice carrying a truncated (Disc1) allele (DN-DISC1). We utilized whole cell recordings to assess electrophysiological properties and modulation by dopamine (DA) in two classes of interneurons: fast-spiking (FS) and low threshold-spiking (LTS) interneurons in wild-type and DN-DISC1 mice. In DN-DISC1 mice, FS interneurons, but not LTS interneurons, exhibited altered action potentials. Further, the perineuronal nets that surround FS interneurons exhibited abnormal morphology in DN-DISC1 mice, and the DA modulation of this cell type was altered in DN-DISC1 mice. We conclude that early-life manipulation of a gene associated with risk of psychiatric disease can result in dysfunction, but not loss, of specific GABAergic interneurons. The resulting alteration of excitatory-inhibitory balance is a critical element in DISC1 pathophysiology.
Collapse
Affiliation(s)
- Ross A Cardarelli
- Program in Neuroscience, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Rolicia Martin
- Department of Anatomy and Neurobiology, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Hanna Jaaro-Peled
- Department of Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Akira Sawa
- Department of Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth M Powell
- Department of Anatomy and Neurobiology, University of Maryland Medical School, Baltimore, Maryland, USA.,Department of Psychiatry, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Patricio O'Donnell
- Program in Neuroscience, University of Maryland Medical School, Baltimore, Maryland, USA.,Department of Anatomy and Neurobiology, University of Maryland Medical School, Baltimore, Maryland, USA.,Department of Psychiatry, University of Maryland Medical School, Baltimore, Maryland, USA
| |
Collapse
|
48
|
Abstract
Axons link distant brain regions and are usually considered as simple transmission cables in which reliable propagation occurs once an action potential has been generated. Safe propagation of action potentials relies on specific ion channel expression at strategic points of the axon such as nodes of Ranvier or axonal branch points. However, while action potentials are generally considered as the quantum of neuronal information, their signaling is not entirely digital. In fact, both their shape and their conduction speed have been shown to be modulated by activity, leading to regulations of synaptic latency and synaptic strength. We report here newly identified mechanisms of (1) safe spike propagation along the axon, (2) compartmentalization of action potential shape in the axon, (3) analog modulation of spike-evoked synaptic transmission and (4) alteration in conduction time after persistent regulation of axon morphology in central neurons. We discuss the contribution of these regulations in information processing.
Collapse
Affiliation(s)
- Sylvain Rama
- UNIS, UMR_S 1072, INSERM, Aix-Marseille Université, 13015 Marseille, France; Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Mickaël Zbili
- UNIS, UMR_S 1072, INSERM, Aix-Marseille Université, 13015 Marseille, France
| | - Dominique Debanne
- UNIS, UMR_S 1072, INSERM, Aix-Marseille Université, 13015 Marseille, France.
| |
Collapse
|
49
|
Sialana FJ, Wang AL, Fazari B, Kristofova M, Smidak R, Trossbach SV, Korth C, Huston JP, de Souza Silva MA, Lubec G. Quantitative Proteomics of Synaptosomal Fractions in a Rat Overexpressing Human DISC1 Gene Indicates Profound Synaptic Dysregulation in the Dorsal Striatum. Front Mol Neurosci 2018; 11:26. [PMID: 29467617 PMCID: PMC5808171 DOI: 10.3389/fnmol.2018.00026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/18/2018] [Indexed: 12/12/2022] Open
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a key protein involved in behavioral processes and various mental disorders, including schizophrenia and major depression. A transgenic rat overexpressing non-mutant human DISC1, modeling aberrant proteostasis of the DISC1 protein, displays behavioral, biochemical and anatomical deficits consistent with aspects of mental disorders, including changes in the dorsal striatum, an anatomical region critical in the development of behavioral disorders. Herein, dorsal striatum of 10 transgenic DISC1 (tgDISC1) and 10 wild type (WT) littermate control rats was used for synaptosomal preparations and for performing liquid chromatography-tandem mass spectrometry (LC-MS)-based quantitative proteomics, using isobaric labeling (TMT10plex). Functional enrichment analysis was generated from proteins with level changes. The increase in DISC1 expression leads to changes in proteins and synaptic-associated processes including membrane trafficking, ion transport, synaptic organization and neurodevelopment. Canonical pathway analysis assigned proteins with level changes to actin cytoskeleton, Gαq, Rho family GTPase and Rho GDI, axonal guidance, ephrin receptor and dopamine-DARPP32 feedback in cAMP signaling. DISC1-regulated proteins proposed in the current study are also highly associated with neurodevelopmental and mental disorders. Bioinformatics analyses from the current study predicted that the following biological processes may be activated by overexpression of DISC1, i.e., regulation of cell quantities, neuronal and axonal extension and long term potentiation. Our findings demonstrate that the effects of overexpression of non-mutant DISC1 or its misassembly has profound consequences on protein networks essential for behavioral control. These results are also relevant for the interpretation of previous as well as for the design of future studies on DISC1.
Collapse
Affiliation(s)
- Fernando J Sialana
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - An-Li Wang
- Center for Behavioral Neuroscience, University of Düsseldorf, Düsseldorf, Germany
| | - Benedetta Fazari
- Center for Behavioral Neuroscience, University of Düsseldorf, Düsseldorf, Germany
| | - Martina Kristofova
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Roman Smidak
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Svenja V Trossbach
- Department of Neuropathology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| | - Carsten Korth
- Department of Neuropathology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany
| | - Joseph P Huston
- Center for Behavioral Neuroscience, University of Düsseldorf, Düsseldorf, Germany
| | | | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Private Medical University, Salzburg, Austria
| |
Collapse
|
50
|
Sultana R, Ghandi T, M. Davila A, Lee CC, Ogundele OM. Upregulated SK2 Expression and Impaired CaMKII Phosphorylation Are Shared Synaptic Defects Between 16p11.2del and 129S: Δdisc1 Mutant Mice. ASN Neuro 2018; 10:1759091418817641. [PMID: 33592687 PMCID: PMC6295693 DOI: 10.1177/1759091418817641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 12/29/2022] Open
Abstract
Ion channel gating and kinase regulation of N-methyl-D-aspartate receptor 1 activity are fundamental mechanisms that govern synaptic plasticity. In this study, we showed that two mutant models (16p11.2del and Δdisc1 ) that recapitulate aspects of human cognitive disorders shared a similar defect in N-methyl-D-aspartate receptor 1-dependent synaptic function. Our results demonstrate that the expression of small-conductance potassium channels (SK2 or KCa2.2) was significantly upregulated in the hippocampus and prefrontal cortex of 16p11.2del and 129S:Δdisc1 mutant mice. Likewise, both mutant strains exhibited an impairment of T286 phosphorylation of calcium-calmodulin-dependent kinase II (CaMKII) in the hippocampus and prefrontal cortex. In vivo neural recordings revealed that increased SK2 expression and impaired T286 phosphorylation of CaMKII coincide with a prolonged interspike interval in the hippocampal cornu ammonis-1 (CA1) field for both 16p11.2del and 129S:Δdisc1 mutant mice. These findings suggest that alteration of small conductance channels and T286 phosphorylation of CaMKII are likely shared factors underlying behavioral changes in these two genetic mouse models.
Collapse
Affiliation(s)
- Razia Sultana
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Tanya Ghandi
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Alexandra M. Davila
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Charles C. Lee
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Olalekan M. Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| |
Collapse
|