201
|
Bijata M, Bączyńska E, Müller FE, Bijata K, Masternak J, Krzystyniak A, Szewczyk B, Siwiec M, Antoniuk S, Roszkowska M, Figiel I, Magnowska M, Olszyński KH, Wardak AD, Hogendorf A, Ruszczycki B, Gorinski N, Labus J, Stępień T, Tarka S, Bojarski AJ, Tokarski K, Filipkowski RK, Ponimaskin E, Wlodarczyk J. Activation of the 5-HT7 receptor and MMP-9 signaling module in the hippocampal CA1 region is necessary for the development of depressive-like behavior. Cell Rep 2022; 38:110532. [PMID: 35294881 DOI: 10.1016/j.celrep.2022.110532] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 10/31/2021] [Accepted: 02/25/2022] [Indexed: 12/13/2022] Open
Abstract
Major depressive disorder is a complex disease resulting from aberrant synaptic plasticity that may be caused by abnormal serotonergic signaling. Using a combination of behavioral, biochemical, and imaging methods, we analyze 5-HT7R/MMP-9 signaling and dendritic spine plasticity in the hippocampus in mice treated with the selective 5-HT7R agonist (LP-211) and in a model of chronic unpredictable stress (CUS)-induced depressive-like behavior. We show that acute 5-HT7R activation induces depressive-like behavior in mice in an MMP-9-dependent manner and that post mortem brain samples from human individuals with depression reveal increased MMP-9 enzymatic activity in the hippocampus. Both pharmacological activation of 5-HT7R and modulation of its downstream effectors as a result of CUS lead to dendritic spine elongation and decreased spine density in this region. Overall, the 5-HT7R/MMP-9 pathway is specifically activated in the CA1 subregion of the hippocampus during chronic stress and is crucial for inducing depressive-like behavior.
Collapse
Affiliation(s)
- Monika Bijata
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland; Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Ewa Bączyńska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland; The Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Franziska E Müller
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Krystian Bijata
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland; Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Julia Masternak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Adam Krzystyniak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Bernadeta Szewczyk
- Maj Institute of Pharmacology, Department of Neurobiology, Polish Academy of Sciences, Smętna 12, 31-343 Cracow, Poland
| | - Marcin Siwiec
- Maj Institute of Pharmacology, Department of Physiology, Polish Academy of Sciences, Smętna 12, 31-343 Cracow, Poland
| | - Svitlana Antoniuk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland; Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Matylda Roszkowska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Izabela Figiel
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Marta Magnowska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Krzysztof H Olszyński
- Behavior and Metabolism Research Laboratory, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Agnieszka D Wardak
- Behavior and Metabolism Research Laboratory, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Adam Hogendorf
- Maj Institute of Pharmacology, Department of Medicinal Chemistry, Polish Academy of Sciences, Smętna 12, 31-343 Cracow, Poland
| | - Błażej Ruszczycki
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland
| | - Nataliya Gorinski
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Josephine Labus
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Tomasz Stępień
- Department of Neuropathology, Institute of Psychiatry and Neurology, Jana III Sobieskiego 9, 02-957 Warsaw, Poland
| | - Sylwia Tarka
- Department of Forensic Medicine, Medical University of Warsaw, Oczki 1, 02-007 Warsaw, Poland
| | - Andrzej J Bojarski
- Maj Institute of Pharmacology, Department of Medicinal Chemistry, Polish Academy of Sciences, Smętna 12, 31-343 Cracow, Poland
| | - Krzysztof Tokarski
- Maj Institute of Pharmacology, Department of Physiology, Polish Academy of Sciences, Smętna 12, 31-343 Cracow, Poland
| | - Robert K Filipkowski
- Behavior and Metabolism Research Laboratory, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Jakub Wlodarczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland.
| |
Collapse
|
202
|
Guzmán Salas S, Weber A, Malci A, Lin X, Herrera-Molina R, Cerpa W, Dorador C, Signorelli J, Zamorano P. The metabolite p-cresol impairs dendritic development, synaptogenesis and synapse function in hippocampal neurons: Implications for autism spectrum disorder. J Neurochem 2022; 161:335-349. [PMID: 35257373 DOI: 10.1111/jnc.15604] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 02/21/2022] [Accepted: 03/01/2022] [Indexed: 11/29/2022]
Abstract
Autism spectrum disorder (ASD) is a heterogenous neurodevelopment disorder resulting from different etiological factors, both genetic and/or environmental. These factors can lead to abnormal neuronal development on dendrite and synaptic function at the central nervous system. Recent studies have shown that a subset of ASD patients display increased circulation levels of the tyrosine metabolite, p-cresol, related to chronic intestinal disorders due to dysbiosis of the intestinal microbiota. In particular, abnormal presence of intestinal Clostridium sp. has been linked to high levels of p-cresol in ASD children younger than 8 years. However, the role of p-cresol during development of the central nervous system is unknown. Here, we evaluated in vitro the effect of p-cresol on neurite outgrowth in N2a and PC12 cell lines and dendritic morphology, synaptic density, neuronal activity, and calcium responses in primary rat hippocampal neurons. p-cresol inhibits neural differentiation and neurites outgrowth in N2a and PC12 neuronal cell lines. In hippocampal neuronal cultures, Sholl´s analysis shows a decrease in the dendritic arborization of neurons treated with p-cresol. Synaptic density analyzed with the synaptic markers Piccolo and Shank2 is diminished in hippocampal neurons treated with p-cresol. Electrically-evoked intracellular calcium rise was drastically, but reversely, blocked by p-cresol, whereas that spontaneous neuronal activity was severely affected by early addition of the metabolite. These findings show that p-cresol alters dendrite development, synaptogenesis and synapse function of neurons in culture, therefore, neuronal alterations occurring in ASD children may be related to this metabolite and dysbiosis of the intestinal microbiota.
Collapse
Affiliation(s)
- Sheyla Guzmán Salas
- Departamento Biomédico, Universidad de Antofagasta, Antofagasta, Chile.,Centre for Biotechnology and Bioengineering CeBiB, Antofagasta
| | - André Weber
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Ayse Malci
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Xiao Lin
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Rodrigo Herrera-Molina
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O'Higgins, Santiago, Chile.,Center for Behavioral Brain Sciences and Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal; Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA); Universidad de Magallanes, Punta Arenas, Chile.,Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristina Dorador
- Centre for Biotechnology and Bioengineering CeBiB, Antofagasta.,Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile
| | | | - Pedro Zamorano
- Departamento Biomédico, Universidad de Antofagasta, Antofagasta, Chile.,Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile
| |
Collapse
|
203
|
Cong Q, Soteros BM, Huo A, Li Y, Tenner AJ, Sia GM. C1q and SRPX2 regulate microglia mediated synapse elimination during early development in the visual thalamus but not the visual cortex. Glia 2022; 70:451-465. [PMID: 34762332 PMCID: PMC8732326 DOI: 10.1002/glia.24114] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/06/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022]
Abstract
The classical complement cascade mediates synapse elimination in the visual thalamus during early brain development. However, whether the primary visual cortex also undergoes complement-mediated synapse elimination during early visual system development remains unknown. Here, we examined microglia-mediated synapse elimination in the visual thalamus and the primary visual cortex of early postnatal C1q and SRPX2 knockout mice. In the lateral geniculate nucleus, deletion of C1q caused a persistent decrease in synapse elimination and microglial synapse engulfment, while deletion of SRPX2 caused a transient increase in the same readouts. In the C1q-SRPX2 double knockout mice, the C1q knockout phenotypes were dominant over the SRPX2 knockout phenotypes, a result which is consistent with SRPX2 being an inhibitor of C1q. We found that genetic deletion of either C1q or SRPX2 did not affect synapse elimination or microglial engulfment of synapses in layer 4 of the primary visual cortex in early brain development. Together, these results show that the classical complement pathway regulates microglia-mediated synapse elimination in the visual thalamus but not the visual cortex during early development of the central nervous system.
Collapse
Affiliation(s)
- Qifei Cong
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA,Institutes of Neuroscience, Soochow University, Suzhou, China.,Corresponding author: ,
| | - Breeanne M. Soteros
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Anran Huo
- Institutes of Neuroscience, Soochow University, Suzhou, China
| | - Yang Li
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Andrea J. Tenner
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA,Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Gek Ming Sia
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA,Corresponding author: ,
| |
Collapse
|
204
|
Yang L, Wu C, Li Y, Dong Y, Wu CYC, Lee RHC, Brann DW, Lin HW, Zhang Q. Long-term exercise pre-training attenuates Alzheimer's disease-related pathology in a transgenic rat model of Alzheimer's disease. GeroScience 2022; 44:1457-1477. [PMID: 35229257 DOI: 10.1007/s11357-022-00534-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Despite enormous efforts around the world, there remains no effective cure for AD. This study was performed to investigate the effects of long-term exercise pretreatment on the typical pathology of AD in a novel transgenic AD rat model. Male 2-month-old animals were divided into the following groups: wild-type (WT) rats, AD rats, and AD rats with treadmill exercise pretreatment (AD-Exe). After exercise pretreatment, the Barnes maze task, passive avoidance task, and cued fear conditioning test were performed to test learning and memory function. The elevated plus maze, open field test, sucrose preference test, and forced swim test were conducted to measure anxious-depressive-like behavior. Immunofluorescence staining, Golgi staining, transmission electron microscopy, Western blot analysis, F-Jade C staining, TUNEL staining, and related assay kits were conducted to measure Aβ plaques, tau hyperphosphorylation, neuronal damage, neuronal degeneration, dendritic spine density, synapses, synaptic vesicles, mitochondrial morphology, mitochondrial dynamic, oxidative stress, and neuroinflammation. Behavioral tests revealed that long-term exercise pretreatment significantly alleviated learning and memory dysfunction and anxious-depressive-like behaviors in AD animals. In addition, exercise pretreatment attenuated amyloid-β deposition and tau hyperphosphorylation and preserved spine density, synapses, and presynaptic vesicles. Exercise also inhibited neuronal damage, neuronal apoptosis, and neuronal degeneration. Additional studies revealed the imbalance of mitochondrial dynamics was significantly inhibited by exercise pretreatment accompanied by a remarkable suppression of oxidative stress and neuroinflammation. Our findings suggest that long-term exercise pretreatment alleviated behavioral deficits and typical pathologies of the AD rat model, supporting long-term exercise pretreatment as a potential approach to delay the progression of AD.
Collapse
Affiliation(s)
- Luodan Yang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Chongyun Wu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yong Li
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Reggie Hui-Chao Lee
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| |
Collapse
|
205
|
Zhu D, Johnson HJ, Chen J, Schaffer DV. Optogenetic Application to Investigating Cell Behavior and Neurological Disease. Front Cell Neurosci 2022; 16:811493. [PMID: 35273478 PMCID: PMC8902366 DOI: 10.3389/fncel.2022.811493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Cells reside in a dynamic microenvironment that presents them with regulatory signals that vary in time, space, and amplitude. The cell, in turn, interprets these signals and accordingly initiates downstream processes including cell proliferation, differentiation, migration, and self-organization. Conventional approaches to perturb and investigate signaling pathways (e.g., agonist/antagonist addition, overexpression, silencing, knockouts) are often binary perturbations that do not offer precise control over signaling levels, and/or provide limited spatial or temporal control. In contrast, optogenetics leverages light-sensitive proteins to control cellular signaling dynamics and target gene expression and, by virtue of precise hardware control over illumination, offers the capacity to interrogate how spatiotemporally varying signals modulate gene regulatory networks and cellular behaviors. Recent studies have employed various optogenetic systems in stem cell, embryonic, and somatic cell patterning studies, which have addressed fundamental questions of how cell-cell communication, subcellular protein localization, and signal integration affect cell fate. Other efforts have explored how alteration of signaling dynamics may contribute to neurological diseases and have in the process created physiologically relevant models that could inform new therapeutic strategies. In this review, we focus on emerging applications within the expanding field of optogenetics to study gene regulation, cell signaling, neurodevelopment, and neurological disorders, and we comment on current limitations and future directions for the growth of the field.
Collapse
Affiliation(s)
- Danqing Zhu
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, United States
| | - Hunter J. Johnson
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, United States
- Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, CA, United States
- Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, United States
| | - Jun Chen
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, United States
| | - David V. Schaffer
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, United States
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, United States
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
- *Correspondence: David V. Schaffer
| |
Collapse
|
206
|
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: 1.5] [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
|
207
|
Terashima H, Minatohara K, Maruoka H, Okabe S. Imaging neural circuit pathology of autism spectrum disorders: autism-associated genes, animal models and the application of in vivo two-photon imaging. Microscopy (Oxf) 2022; 71:i81-i99. [DOI: 10.1093/jmicro/dfab039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/11/2021] [Accepted: 11/08/2021] [Indexed: 11/12/2022] Open
Abstract
Abstract
Recent advances in human genetics identified genetic variants involved in causing autism spectrum disorders (ASDs). Mouse models that mimic mutations found in patients with ASD exhibit behavioral phenotypes consistent with ASD symptoms. These mouse models suggest critical biological factors of ASD etiology. Another important implication of ASD genetics is the enrichment of ASD risk genes in molecules involved in developing synapses and regulating neural circuit function. Sophisticated in vivo imaging technologies applied to ASD mouse models identify common synaptic impairments in the neocortex, with genetic-mutation-specific defects in local neural circuits. In this article, we review synapse- and circuit-level phenotypes identified by in vivo two-photon imaging in multiple mouse models of ASD and discuss the contributions of altered synapse properties and neural circuit activity to ASD pathogenesis.
Collapse
Affiliation(s)
- Hiroshi Terashima
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Keiichiro Minatohara
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hisato Maruoka
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
208
|
Al Dera H. Cellular and molecular mechanisms underlying autism spectrum disorders and associated comorbidities: A pathophysiological review. Biomed Pharmacother 2022; 148:112688. [PMID: 35149383 DOI: 10.1016/j.biopha.2022.112688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/25/2022] [Accepted: 02/01/2022] [Indexed: 12/31/2022] Open
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders that develop in early life due to interaction between several genetic and environmental factors and lead to alterations in brain function and structure. During the last decades, several mechanisms have been placed to explain the pathogenesis of autism. Unfortunately, these are reported in several studies and reviews which make it difficult to follow by the reader. In addition, some recent molecular mechanisms related to ASD have been unrevealed. This paper revises and highlights the major common molecular mechanisms responsible for the clinical symptoms seen in people with ASD, including the roles of common genetic factors and disorders, neuroinflammation, GABAergic signaling, and alterations in Ca+2 signaling. Besides, it covers the major molecular mechanisms and signaling pathways involved in initiating the epileptic seizure, including the alterations in the GABAergic and glutamate signaling, vitamin and mineral deficiency, disorders of metabolism, and autoimmunity. Finally, this review also discusses sleep disorder patterns and the molecular mechanisms underlying them.
Collapse
Affiliation(s)
- Hussain Al Dera
- Department of Basic Medical Sciences, College of Medicine at King Saud, Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia.
| |
Collapse
|
209
|
Vanderplow AM, Kermath BA, Bernhardt CR, Gums KT, Seablom EN, Radcliff AB, Ewald AC, Jones MV, Baker TL, Watters JJ, Cahill ME. A feature of maternal sleep apnea during gestation causes autism-relevant neuronal and behavioral phenotypes in offspring. PLoS Biol 2022; 20:e3001502. [PMID: 35113852 PMCID: PMC8812875 DOI: 10.1371/journal.pbio.3001502] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/29/2021] [Indexed: 12/26/2022] Open
Abstract
Mounting epidemiologic and scientific evidence indicates that many psychiatric disorders originate from a complex interplay between genetics and early life experiences, particularly in the womb. Despite decades of research, our understanding of the precise prenatal and perinatal experiences that increase susceptibility to neurodevelopmental disorders remains incomplete. Sleep apnea (SA) is increasingly common during pregnancy and is characterized by recurrent partial or complete cessations in breathing during sleep. SA causes pathological drops in blood oxygen levels (intermittent hypoxia, IH), often hundreds of times each night. Although SA is known to cause adverse pregnancy and neonatal outcomes, the long-term consequences of maternal SA during pregnancy on brain-based behavioral outcomes and associated neuronal functioning in the offspring remain unknown. We developed a rat model of maternal SA during pregnancy by exposing dams to IH, a hallmark feature of SA, during gestational days 10 to 21 and investigated the consequences on the offspring's forebrain synaptic structure, synaptic function, and behavioral phenotypes across multiples stages of development. Our findings represent a rare example of prenatal factors causing sexually dimorphic behavioral phenotypes associated with excessive (rather than reduced) synapse numbers and implicate hyperactivity of the mammalian target of rapamycin (mTOR) pathway in contributing to the behavioral aberrations. These findings have implications for neuropsychiatric disorders typified by superfluous synapse maintenance that are believed to result, at least in part, from largely unknown insults to the maternal environment.
Collapse
Affiliation(s)
- Amanda M. Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Bailey A. Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Cassandra R. Bernhardt
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kimberly T. Gums
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Erin N. Seablom
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Abigail B. Radcliff
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Andrea C. Ewald
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mathew V. Jones
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Tracy L. Baker
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jyoti J. Watters
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michael E. Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
210
|
Fei E, Chen P, Zhang Q, Zhong Y, Zhou T. Protein kinase B/Akt1 phosphorylates dysbindin-1A at serine 10 to regulate neuronal development. Neuroscience 2022; 490:66-78. [DOI: 10.1016/j.neuroscience.2022.01.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 01/05/2023]
|
211
|
Li Y, Ma C, Li S, Wang J, Li W, Yang Y, Li X, Liu J, Yang J, Liu Y, Li K, Li J, Huang D, Chen R, Lv L, Xiao X, Li M, Luo X. Regulatory Variant rs2535629 in ITIH3 Intron Confers Schizophrenia Risk By Regulating CTCF Binding and SFMBT1 Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104786. [PMID: 34978167 PMCID: PMC8867204 DOI: 10.1002/advs.202104786] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/16/2021] [Indexed: 06/14/2023]
Abstract
Genome-wide association studies have identified 3p21.1 as a robust risk locus for schizophrenia. However, the underlying molecular mechanisms remain elusive. Here a functional regulatory variant (rs2535629) is identified that disrupts CTCF binding at 3p21.1. It is confirmed that rs2535629 is also significantly associated with schizophrenia in Chinese population and the regulatory effect of rs2535629 is validated. Expression quantitative trait loci analysis indicates that rs2535629 is associated with the expression of three distal genes (GLT8D1, SFMBT1, and NEK4) in the human brain, and CRISPR-Cas9-mediated genome editing confirmed the regulatory effect of rs2535629 on GLT8D1, SFMBT1, and NEK4. Interestingly, differential expression analysis of GLT8D1, SFMBT1, and NEK4 suggested that rs2535629 may confer schizophrenia risk by regulating SFMBT1 expression. It is further demonstrated that Sfmbt1 regulates neurodevelopment and dendritic spine density, two key pathological characteristics of schizophrenia. Transcriptome analysis also support the potential role of Sfmbt1 in schizophrenia pathogenesis. The study identifies rs2535629 as a plausibly causal regulatory variant at the 3p21.1 risk locus and demonstrates the regulatory mechanism and biological effect of this functional variant, indicating that this functional variant confers schizophrenia risk by altering CTCF binding and regulating expression of SFMBT1, a distal gene which plays important roles in neurodevelopment and synaptic morphogenesis.
Collapse
Affiliation(s)
- Yifan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Changguo Ma
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Yunnan Key Laboratory for Basic Research on Bone and Joint Diseases & Yunnan Stem Cell Translational Research CenterKunming UniversityKunmingYunnan650214China
| | - Shiwu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Junyang Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Wenqiang Li
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangHenan453002China
- Henan Key Lab of Biological PsychiatryInternational Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangHenan453002China
| | - Yongfeng Yang
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangHenan453002China
- Henan Key Lab of Biological PsychiatryInternational Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangHenan453002China
| | - Xiaoyan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of EducationInstitutes of Physical Science and Information TechnologyAnhui UniversityHefeiAnhui230601China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
| | - Jinfeng Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Yixing Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Kaiqin Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Jiao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Di Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
| | - Rui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
| | - Luxian Lv
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangHenan453002China
- Henan Key Lab of Biological PsychiatryInternational Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangHenan453002China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
| | - Xiong‐Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingYunnan650204China
- Center for Excellence in Animal Evolution and GeneticsChinese Academy of SciencesKunmingYunnan650204China
- KIZ‐CUHK Joint Laboratory of Bioresources and Molecular Research in Common DiseasesKunming Institute of ZoologyChinese Academy of SciencesKunmingYunnan650204China
| |
Collapse
|
212
|
Analysis of mRNA and Protein Levels of CAP2, DLG1 and ADAM10 Genes in Post-Mortem Brain of Schizophrenia, Parkinson's and Alzheimer's Disease Patients. Int J Mol Sci 2022; 23:ijms23031539. [PMID: 35163460 PMCID: PMC8835961 DOI: 10.3390/ijms23031539] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 12/22/2022] Open
Abstract
Schizophrenia (SCZ) is a mental illness characterized by aberrant synaptic plasticity and connectivity. A large bulk of evidence suggests genetic and functional links between postsynaptic abnormalities and SCZ. Here, we performed quantitative PCR and Western blotting analysis in the dorsolateral prefrontal cortex (DLPFC) and hippocampus of SCZ patients to investigate the mRNA and protein expression of three key spine shapers: the actin-binding protein cyclase-associated protein 2 (CAP2), the sheddase a disintegrin and metalloproteinase 10 (ADAM10), and the synapse-associated protein 97 (SAP97). Our analysis of the SCZ post-mortem brain indicated increased DLG1 mRNA in DLPFC and decreased CAP2 mRNA in the hippocampus of SCZ patients, compared to non-psychiatric control subjects, while the ADAM10 transcript was unaffected. Conversely, no differences in CAP2, SAP97, and ADAM10 protein levels were detected between SCZ and control individuals in both brain regions. To assess whether DLG1 and CAP2 transcript alterations were selective for SCZ, we also measured their expression in the superior frontal gyrus of patients affected by neurodegenerative disorders, like Parkinson’s and Alzheimer’s disease. Interestingly, also in Parkinson’s disease patients, we found a selective reduction of CAP2 mRNA levels relative to controls but unaltered protein levels. Taken together, we reported for the first time altered CAP2 expression in the brain of patients with psychiatric and neurological disorders, thus suggesting that aberrant expression of this gene may contribute to synaptic dysfunction in these neuropathologies.
Collapse
|
213
|
[Effects of ring finger and tryptophan-aspartic acid 2 on dendritic spines and synapse formation in cerebral cortex neurons of mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:78-85. [PMID: 35249873 PMCID: PMC8901395 DOI: 10.12122/j.issn.1673-4254.2022.01.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To clarify the functional effects of differential expression of ring finger and tryptophan-aspartic acid 2 (RFWD2) on dendritic development and formation of dendritic spines in cerebral cortex neurons of mice. METHODS Immunofluorescent staining was used to identify the location and global expression profile of RFWD2 in mouse brain and determine the co-localization of RFWD2 with the synaptic proteins in the cortical neurons. We also examined the effects of RFWD2 over-expression (RFWD2-Myc) and RFWD2 knockdown (RFWD2-shRNA) on dendritic development, dendritic spine formation and synaptic function in cultured cortical neurons. RESULTS RFWD2 is highly expressed in the cerebral cortex and hippocampus of mice, and its expression level was positively correlated with the development of cerebral cortex neurons and dendrites. RFWD2 expression was detected on the presynaptic membrane and postsynaptic membrane of the neurons, and its expression levels were positively correlated with the length, number of branches and complexity of the dendrites. In cultured cortical neurons, RFWD2 overexpression significantly lowered the expressions of the synaptic proteins synaptophysin (P < 0.01) and postsynapic density protein 95 (P < 0.01), while RFWD2 knockdown significantly increased their expressions (both P < 0.05). Compared with the control and RFWD2-overexpressing cells, the neurons with RFWD2 knockdown showed significantly reduced number of dendritic spines (both P < 0.05). CONCLUSION RFWD2 can regulate the expression of the synaptic proteins, the development of the dendrites, the formation of the dendritic spines and synaptic function in mouse cerebral cortex neurons through ubiquitination of Pea3 family members and c-Jun, which may serve as potential treatment targets for neurological diseases.
Collapse
|
214
|
Perna J, Lu J, Mullen B, Liu T, Tjia M, Weiser S, Ackman J, Zuo Y. Perinatal Penicillin Exposure Affects Cortical Development and Sensory Processing. Front Mol Neurosci 2022; 14:704219. [PMID: 35002614 PMCID: PMC8727458 DOI: 10.3389/fnmol.2021.704219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 11/22/2021] [Indexed: 12/19/2022] Open
Abstract
The prevalent use of antibiotics in pregnant women and neonates raises concerns about long-term risks for children’s health, but their effects on the central nervous system is not well understood. We studied the effects of perinatal penicillin exposure (PPE) on brain structure and function in mice with a therapeutically relevant regimen. We used a battery of behavioral tests to evaluate anxiety, working memory, and sensory processing, and immunohistochemistry to quantify changes in parvalbumin-expressing inhibitory interneurons (PV+ INs), perineuronal nets (PNNs), as well as microglia density and morphology. In addition, we performed mesoscale calcium imaging to study neural activity and functional connectivity across cortical regions, and two-photon imaging to monitor dendritic spine and microglial dynamics. We found that adolescent PPE mice have abnormal sensory processing, including impaired texture discrimination and altered prepulse inhibition. Such behavioral changes are associated with increased spontaneous neural activities in various cortical regions, and delayed maturation of PV+ INs in the somatosensory cortex. Furthermore, adolescent PPE mice have elevated elimination of dendritic spines on the apical dendrites of layer 5 pyramidal neurons, as well as increased ramifications and spatial coverage of cortical microglia. Finally, while synaptic defects are transient during adolescence, behavioral abnormalities persist into adulthood. Our study demonstrates that early-life exposure to antibiotics affects cortical development, leaving a lasting effect on brain functions.
Collapse
Affiliation(s)
- James Perna
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Ju Lu
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Brian Mullen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Taohui Liu
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Michelle Tjia
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Sydney Weiser
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - James Ackman
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
215
|
Renna P, Ripoli C, Dagliyan O, Pastore F, Rinaudo M, Re A, Paciello F, Grassi C. Engineering a switchable single‐chain
TEV
protease to control protein maturation in living neurons. Bioeng Transl Med 2022; 7:e10292. [PMID: 35600650 PMCID: PMC9115699 DOI: 10.1002/btm2.10292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/13/2021] [Accepted: 12/30/2021] [Indexed: 11/18/2022] Open
Abstract
Engineered proteases are promising tools to address physiological and pathophysiological questions as well as to develop new therapeutic approaches. Here we introduce a new genetically encoded engineered single‐chain tobacco etch virus protease, allowing to control proprotein cleavage in different compartments of living mammalian cells. We demonstrated a set of controllable proteolytic effects, including cytosolic protein cleavage, inducible gene expression, and maturation of brain‐derived neurotrophic factor (BDNF) in the secretory pathway thus showing the versatility of this technique. Of note, the secretory pathway exhibits different characteristics from the cytosol and it is difficult to target because inaccessible to some small molecules. We were able to induce ligand‐mediated BDNF maturation and monitor its effects on dendritic spines in hippocampal pyramidal cells and in the mouse brain. This strategy paves the way to dissect proteolytic cleavage product signaling in various processes as well as for future therapeutic applications.
Collapse
Affiliation(s)
- Pietro Renna
- Department of Neuroscience Università Cattolica del Sacro Cuore, 00168 Rome Italy
| | - Cristian Ripoli
- Department of Neuroscience Università Cattolica del Sacro Cuore, 00168 Rome Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome Italy
| | - Onur Dagliyan
- Department of Neurobiology Harvard Medical School Boston MA USA
| | - Francesco Pastore
- Department of Neuroscience Università Cattolica del Sacro Cuore, 00168 Rome Italy
| | - Marco Rinaudo
- Department of Neuroscience Università Cattolica del Sacro Cuore, 00168 Rome Italy
| | - Agnese Re
- Department of Neuroscience Università Cattolica del Sacro Cuore, 00168 Rome Italy
| | - Fabiola Paciello
- Department of Neuroscience Università Cattolica del Sacro Cuore, 00168 Rome Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome Italy
| | - Claudio Grassi
- Department of Neuroscience Università Cattolica del Sacro Cuore, 00168 Rome Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome Italy
| |
Collapse
|
216
|
Zhang CY, Xiao X, Zhang Z, Hu Z, Li M. An alternative splicing hypothesis for neuropathology of schizophrenia: evidence from studies on historical candidate genes and multi-omics data. Mol Psychiatry 2022; 27:95-112. [PMID: 33686213 DOI: 10.1038/s41380-021-01037-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 01/08/2021] [Accepted: 01/22/2021] [Indexed: 01/31/2023]
Abstract
Alternative splicing of schizophrenia risk genes, such as DRD2, GRM3, and DISC1, has been extensively described. Nevertheless, the alternative splicing characteristics of the growing number of schizophrenia risk genes identified through genetic analyses remain relatively opaque. Recently, transcriptomic analyses in human brains based on short-read RNA-sequencing have discovered many "local splicing" events (e.g., exon skipping junctions) associated with genetic risk of schizophrenia, and further molecular characterizations have identified novel spliced isoforms, such as AS3MTd2d3 and ZNF804AE3E4. In addition, long-read sequencing analyses of schizophrenia risk genes (e.g., CACNA1C and NRXN1) have revealed multiple previously unannotated brain-abundant isoforms with therapeutic potentials, and functional analyses of KCNH2-3.1 and Ube3a1 have provided examples for investigating such spliced isoforms in vitro and in vivo. These findings suggest that alternative splicing may be an essential molecular mechanism underlying genetic risk of schizophrenia, however, the incomplete annotations of human brain transcriptomes might have limited our understanding of schizophrenia pathogenesis, and further efforts to elucidate these transcriptional characteristics are urgently needed to gain insights into the illness-correlated brain physiology and pathology as well as to translate genetic discoveries into novel therapeutic targets.
Collapse
Affiliation(s)
- Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhuohua Zhang
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Zhonghua Hu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China. .,Eye Center of Xiangya Hospital and Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, Hunan, China. .,National Clinical Research Center on Mental Disorders, Changsha, Hunan, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China. .,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
217
|
Hu C, Li H, Li J, Luo X, Hao Y. Microglia: Synaptic modulator in autism spectrum disorder. Front Psychiatry 2022; 13:958661. [PMID: 36465285 PMCID: PMC9714329 DOI: 10.3389/fpsyt.2022.958661] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/28/2022] [Indexed: 11/18/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by variable impairment of social communication and repetitive behaviors, highly restricted interests, and/or sensory behaviors beginning early in life. Many individuals with ASD have dysfunction of microglia, which may be closely related to neuroinflammation, making microglia play an important role in the pathogenesis of ASD. Mounting evidence indicates that microglia, the resident immune cells of the brain, are required for proper brain function, especially in the maintenance of neuronal circuitry and control of behavior. Dysfunction of microglia will ultimately affect the neural function in a variety of ways, including the formation of synapses and alteration of excitatory-inhibitory balance. In this review, we provide an overview of how microglia actively interact with neurons in physiological conditions and modulate the fate and functions of synapses. We put a spotlight on the multi-dimensional neurodevelopmental roles of microglia, especially in the essential influence of synapses, and discuss how microglia are currently thought to influence ASD progression.
Collapse
Affiliation(s)
- Cong Hu
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heli Li
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinhui Li
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Hao
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
218
|
Tanaka A, Inami W, Suzuki Y, Kawata Y. Development of a direct point electron beam exposure system to investigate the biological functions of subcellular domains in a living biological cell. Micron 2022; 155:103214. [DOI: 10.1016/j.micron.2022.103214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 11/26/2022]
|
219
|
Garrido D, Beretta S, Grabrucker S, Bauer HF, Bayer D, Sala C, Verpelli C, Roselli F, Bockmann J, Proepper C, Catanese A, Boeckers TM. Shank2/3 double knockout-based screening of cortical subregions links the retrosplenial area to the loss of social memory in autism spectrum disorders. Mol Psychiatry 2022; 27:4994-5006. [PMID: 36100669 PMCID: PMC9763120 DOI: 10.1038/s41380-022-01756-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 08/15/2022] [Accepted: 08/15/2022] [Indexed: 01/19/2023]
Abstract
Members of the Shank protein family are master scaffolds of the postsynaptic architecture and mutations within the SHANK genes are causally associated with autism spectrum disorders (ASDs). We generated a Shank2-Shank3 double knockout mouse that is showing severe autism related core symptoms, as well as a broad spectrum of comorbidities. We exploited this animal model to identify cortical brain areas linked to specific autistic traits by locally deleting Shank2 and Shank3 simultaneously. Our screening of 10 cortical subregions revealed that a Shank2/3 deletion within the retrosplenial area severely impairs social memory, a core symptom of ASD. Notably, DREADD-mediated neuronal activation could rescue the social impairment triggered by Shank2/3 depletion. Data indicate that the retrosplenial area has to be added to the list of defined brain regions that contribute to the spectrum of behavioural alterations seen in ASDs.
Collapse
Affiliation(s)
- Débora Garrido
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany ,grid.6582.90000 0004 1936 9748International Graduate School, Ulm University, 89081 Ulm, Germany
| | - Stefania Beretta
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Ulm site, 89081 Ulm, Germany
| | - Stefanie Grabrucker
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Helen Friedericke Bauer
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany ,grid.6582.90000 0004 1936 9748International Graduate School, Ulm University, 89081 Ulm, Germany
| | - David Bayer
- grid.6582.90000 0004 1936 9748International Graduate School, Ulm University, 89081 Ulm, Germany ,grid.6582.90000 0004 1936 9748Department of Neurology, Ulm University, 89081 Ulm, Germany
| | - Carlo Sala
- grid.418879.b0000 0004 1758 9800CNR, Institute for Neuroscience, Milano, Italy
| | - Chiara Verpelli
- grid.418879.b0000 0004 1758 9800CNR, Institute for Neuroscience, Milano, Italy
| | - Francesco Roselli
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Ulm site, 89081 Ulm, Germany ,grid.6582.90000 0004 1936 9748Department of Neurology, Ulm University, 89081 Ulm, Germany
| | - Juergen Bockmann
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Christian Proepper
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Alberto Catanese
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Ulm site, 89081 Ulm, Germany
| | - Tobias M. Boeckers
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Ulm site, 89081 Ulm, Germany
| |
Collapse
|
220
|
Park Y, Singh P, Fai TG. Coarse-grained Stochastic Model of Myosin-Driven Vesicles into Dendritic Spines. SIAM JOURNAL ON APPLIED MATHEMATICS 2022; 82:793-820. [PMID: 36314039 PMCID: PMC9603279 DOI: 10.1137/21m1434180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
We study the dynamics of membrane vesicle motor transport into dendritic spines, which are bulbous intracellular compartments in neurons that play a key role in transmitting signals between neurons. We consider the stochastic analog of the vesicle transport model in [Park and Fai, The Dynamics of Vesicles Driven Into Closed Constrictions by Molecular Motors. Bull. Math. Biol. 82, 141 (2020)]. The stochastic version, which may be considered as an agent-based model, relies mostly on the action of individual myosin motors to produce vesicle motion. To aid in our analysis, we coarse-grain this agent-based model using a master equation combined with a partial differential equation describing the probability of local motor positions. We confirm through convergence studies that the coarse-graining captures the essential features of bistability in velocity (observed in experiments) and waiting-time distributions to switch between steady-state velocities. Interestingly, these results allow us to reformulate the translocation problem in terms of conditional mean first passage times for a run-and-tumble particle moving on a finite domain with absorbing boundaries at the two ends. We conclude by presenting numerical and analytical calculations of vesicle translocation.
Collapse
Affiliation(s)
- Youngmin Park
- Department of Mathematics, Brandeis University, Waltham, MA 02453, USA
- Corresponding author
| | - Prashant Singh
- International Centre for Theoretical Sciences, TIFR, Bengaluru 560089, India
| | - Thomas G. Fai
- Department of Mathematics, Brandeis University, Waltham, MA 02453, USA
- Volen Center for Complex Systems, Waltham, MA 02453, USA
| |
Collapse
|
221
|
Yan Y, Tian M, Li M, Zhou G, Chen Q, Xu M, Hu Y, Luo W, Guo X, Zhang C, Xie H, Wu QF, Xiong W, Liu S, Guan JS. ASH1L haploinsufficiency results in autistic-like phenotypes in mice and links Eph receptor gene to autism spectrum disorder. Neuron 2022; 110:1156-1172.e9. [DOI: 10.1016/j.neuron.2021.12.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/23/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022]
|
222
|
Antunes ASLM, Saia-Cereda VM, Crunfli F, Martins-de-Souza D. 14-3-3 proteins at the crossroads of neurodevelopment and schizophrenia. World J Biol Psychiatry 2022; 23:14-32. [PMID: 33952049 DOI: 10.1080/15622975.2021.1925585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The 14-3-3 family comprises multifunctional proteins that play a role in neurogenesis, neuronal migration, neuronal differentiation, synaptogenesis and dopamine synthesis. 14-3-3 members function as adaptor proteins and impact a wide variety of cellular and physiological processes involved in the pathophysiology of neurological disorders. Schizophrenia is a psychiatric disorder and knowledge about its pathophysiology is still limited. 14-3-3 have been proven to be linked with the dopaminergic, glutamatergic and neurodevelopmental hypotheses of schizophrenia. Further, research using genetic models has demonstrated the role played by 14-3-3 proteins in neurodevelopment and neuronal circuits, however a more integrative and comprehensive approach is needed for a better understanding of their role in schizophrenia. For instance, we still lack an integrated assessment of the processes affected by 14-3-3 proteins in the dopaminergic and glutamatergic systems. In this context, it is also paramount to understand their involvement in the biology of brain cells other than neurons. Here, we present previous and recent research that has led to our current understanding of the roles 14-3-3 proteins play in brain development and schizophrenia, perform an assessment of their functional protein association network and discuss the use of protein-protein interaction modulators to target 14-3-3 as a potential therapeutic strategy.
Collapse
Affiliation(s)
- André S L M Antunes
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Verônica M Saia-Cereda
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Fernanda Crunfli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas, Campinas, Brazil.,Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, SP, Brazil.,D'Or Institute for Research and Education (IDOR), São Paulo, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
| |
Collapse
|
223
|
AUTS2 Gene: Keys to Understanding the Pathogenesis of Neurodevelopmental Disorders. Cells 2021; 11:cells11010011. [PMID: 35011572 PMCID: PMC8750789 DOI: 10.3390/cells11010011] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/08/2021] [Accepted: 12/18/2021] [Indexed: 01/01/2023] Open
Abstract
Neurodevelopmental disorders (NDDs), including autism spectrum disorders (ASD) and intellectual disability (ID), are a large group of neuropsychiatric illnesses that occur during early brain development, resulting in a broad spectrum of syndromes affecting cognition, sociability, and sensory and motor functions. Despite progress in the discovery of various genetic risk factors thanks to the development of novel genomics technologies, the precise pathological mechanisms underlying the onset of NDDs remain elusive owing to the profound genetic and phenotypic heterogeneity of these conditions. Autism susceptibility candidate 2 (AUTS2) has emerged as a crucial gene associated with a wide range of neuropsychological disorders, such as ASD, ID, schizophrenia, and epilepsy. AUTS2 has been shown to be involved in multiple neurodevelopmental processes; in cell nuclei, it acts as a key transcriptional regulator in neurodevelopment, whereas in the cytoplasm, it participates in cerebral corticogenesis, including neuronal migration and neuritogenesis, through the control of cytoskeletal rearrangements. Postnatally, AUTS2 regulates the number of excitatory synapses to maintain the balance between excitation and inhibition in neural circuits. In this review, we summarize the knowledge regarding AUTS2, including its molecular and cellular functions in neurodevelopment, its genetics, and its role in behaviors.
Collapse
|
224
|
Huber N, Hoffmann D, Giniatullina R, Rostalski H, Leskelä S, Takalo M, Natunen T, Solje E, Remes AM, Giniatullin R, Hiltunen M, Haapasalo A. C9orf72 hexanucleotide repeat expansion leads to altered neuronal and dendritic spine morphology and synaptic dysfunction. Neurobiol Dis 2021; 162:105584. [PMID: 34915153 DOI: 10.1016/j.nbd.2021.105584] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/26/2021] [Accepted: 12/11/2021] [Indexed: 12/14/2022] Open
Abstract
Frontotemporal lobar degeneration (FTLD) comprises a heterogenous group of progressive neurodegenerative syndromes. To date, no validated biomarkers or effective disease-modifying therapies exist for the different clinical or genetic subtypes of FTLD. The most common genetic cause underlying FTLD and amyotrophic lateral sclerosis (ALS) is a hexanucleotide repeat expansion in the C9orf72 gene (C9-HRE). FTLD is accompanied by changes in several neurotransmitter systems, including the glutamatergic, GABAergic, dopaminergic, and serotonergic systems and many clinical symptoms can be explained by disturbances in these systems. Here, we aimed to elucidate the effects of the C9-HRE on synaptic function, molecular composition of synapses, and dendritic spine morphology. We overexpressed the pathological C9-HRE in cultured E18 mouse primary hippocampal neurons and characterized the pathological, morphological, and functional changes by biochemical methods, confocal microscopy, and live cell calcium imaging. The C9-HRE-expressing neurons were confirmed to display the pathological RNA foci and DPR proteins. C9-HRE expression led to significant changes in dendritic spine morphologies, as indicated by decreased number of mushroom-type spines and increased number of stubby and thin spines, as well as diminished neuronal branching. These morphological changes were accompanied by concomitantly enhanced susceptibility of the neurons to glutamate-induced excitotoxicity as well as augmented and prolonged responses to excitatory stimuli by glutamate and depolarizing potassium chloride as compared to control neurons. Mechanistically, the hyperexcitation phenotype in the C9-HRE-expressing neurons was found to be underlain by increased activity of extrasynaptic GluN2B-containing N-methyl-d-aspartate (NMDA) receptors. Our results are in accordance with the idea suggesting that C9-HRE is associated with enhanced excitotoxicity and synaptic dysfunction. Thus, therapeutic interventions targeted to alleviate synaptic disturbances might offer efficient avenues for the treatment of patients with C9-HRE-associated FTLD.
Collapse
Affiliation(s)
- Nadine Huber
- Molecular Neurodegeneration group, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland.
| | - Dorit Hoffmann
- Molecular Neurodegeneration group, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland.
| | - Raisa Giniatullina
- Molecular Pain Research group, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland.
| | - Hannah Rostalski
- Molecular Neurodegeneration group, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland.
| | - Stina Leskelä
- Molecular Neurodegeneration group, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland.
| | - Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, Yliopistonranta 1E, 70211 Kuopio, Finland.
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, Yliopistonranta 1E, 70211 Kuopio, Finland.
| | - Eino Solje
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland; Neuro Center, Neurology, Kuopio University Hospital, P. O. Box 100, FI-70029 KYS, Finland.
| | - Anne M Remes
- Medical Research Center, Oulu University Hospital, P. O. Box 8000, FI-90014 University of Oulu, Finland; Unit of Clinical Neuroscience, Neurology, University of Oulu, P. O. Box 8000, FI-90014 University of Oulu, Finland.
| | - Rashid Giniatullin
- Molecular Pain Research group, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland.
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Yliopistonranta 1E, 70211 Kuopio, Finland.
| | - Annakaisa Haapasalo
- Molecular Neurodegeneration group, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland.
| |
Collapse
|
225
|
Musi CA, Castaldo AM, Valsecchi AE, Cimini S, Morello N, Pizzo R, Renieri A, Meloni I, Bonati M, Giustetto M, Borsello T. JNK signaling provides a novel therapeutic target for Rett syndrome. BMC Biol 2021; 19:256. [PMID: 34911542 PMCID: PMC8675514 DOI: 10.1186/s12915-021-01190-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/11/2021] [Indexed: 11/24/2022] Open
Abstract
Background Rett syndrome (RTT) is a monogenic X-linked neurodevelopmental disorder characterized by loss-of-function mutations in the MECP2 gene, which lead to structural and functional changes in synapse communication, and impairments of neural activity at the basis of cognitive deficits that progress from an early age. While the restoration of MECP2 in animal models has been shown to rescue some RTT symptoms, gene therapy intervention presents potential side effects, and with gene- and RNA-editing approaches still far from clinical application, strategies focusing on signaling pathways downstream of MeCP2 may provide alternatives for the development of more effective therapies in vivo. Here, we investigate the role of the c-Jun N-terminal kinase (JNK) stress pathway in the pathogenesis of RTT using different animal and cell models and evaluate JNK inhibition as a potential therapeutic approach. Results We discovered that the c-Jun N-terminal kinase (JNK) stress pathway is activated in Mecp2-knockout, Mecp2-heterozygous mice, and in human MECP2-mutated iPSC neurons. The specific JNK inhibitor, D-JNKI1, promotes recovery of body weight and locomotor impairments in two mouse models of RTT and rescues their dendritic spine alterations. Mecp2-knockout presents intermittent crises of apnea/hypopnea, one of the most invalidating RTT pathological symptoms, and D-JNKI1 powerfully reduces this breathing dysfunction. Importantly, we discovered that also neurons derived from hiPSC-MECP2 mut show JNK activation, high-phosphorylated c-Jun levels, and cell death, which is not observed in the isogenic control wt allele hiPSCs. Treatment with D-JNKI1 inhibits neuronal death induced by MECP2 mutation in hiPSCs mut neurons. Conclusions As a summary, we found altered JNK signaling in models of RTT and suggest that D-JNKI1 treatment prevents clinical symptoms, with coherent results at the cellular, molecular, and functional levels. This is the first proof of concept that JNK plays a key role in RTT and its specific inhibition offers a new and potential therapeutic tool to tackle RTT. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01190-2.
Collapse
Affiliation(s)
- Clara Alice Musi
- Department of Pharmacological and Biomolecular Sciences, Milan University, Via Balzaretti 9, 20133, Milan, Italy.,Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Anna Maria Castaldo
- Department of Pharmacological and Biomolecular Sciences, Milan University, Via Balzaretti 9, 20133, Milan, Italy
| | | | - Sara Cimini
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Noemi Morello
- Department of Neuroscience and National Institute of Neuroscience, University of Turin, Turin, Italy
| | - Riccardo Pizzo
- Department of Neuroscience and National Institute of Neuroscience, University of Turin, Turin, Italy
| | | | | | - Maurizio Bonati
- Department of Public Heath, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Maurizio Giustetto
- Department of Neuroscience and National Institute of Neuroscience, University of Turin, Turin, Italy
| | - Tiziana Borsello
- Department of Pharmacological and Biomolecular Sciences, Milan University, Via Balzaretti 9, 20133, Milan, Italy. .,Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Via Mario Negri 2, 20156, Milan, Italy.
| |
Collapse
|
226
|
Sethi S, Keil Stietz KP, Valenzuela AE, Klocke CR, Silverman JL, Puschner B, Pessah IN, Lein PJ. Developmental Exposure to a Human-Relevant Polychlorinated Biphenyl Mixture Causes Behavioral Phenotypes That Vary by Sex and Genotype in Juvenile Mice Expressing Human Mutations That Modulate Neuronal Calcium. Front Neurosci 2021; 15:766826. [PMID: 34938155 PMCID: PMC8685320 DOI: 10.3389/fnins.2021.766826] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/17/2021] [Indexed: 01/13/2023] Open
Abstract
Polychlorinated biphenyls (PCBs) are putative environmental risks for neurodevelopmental disorders. Here, we tested two hypotheses: (1) developmental exposure to a human-relevant PCB mixture causes behavioral phenotypes relevant to neurodevelopmental disorders; and (2) expression of human mutations that dysregulate neuronal Ca2+ homeostasis influence sensitivity to behavioral effects of developmental PCB exposures. To test these hypotheses, we used mice that expressed a gain-of-function mutation (T4826I) in ryanodine receptor 1 (RYR1), the X-linked fragile X mental retardation 1 (FMR1) CGG repeat expansion or both mutations (double mutant; DM). Transgenic mice and wildtype (WT) mice were exposed to the MARBLES PCB mix at 0, 0.1, 1, and 6 mg/kg/day in the maternal diet throughout gestation and lactation. The MARBLES PCB mix simulates the relative proportions of the 12 most abundant PCB congeners found in the serum of pregnant women at increased risk for having a child with a neurodevelopmental disorder. We assessed ultrasonic vocalizations at postnatal day 7 (P7), spontaneous repetitive behaviors at P25-P30, and sociability at P27-P32. Developmental PCB exposure reduced ultrasonic vocalizations in WT litters in all dose groups, but had no effect on ultrasonic vocalizations in transgenic litters. Developmental PCB exposure significantly increased self-grooming and decreased sociability in WT males in the 0.1 mg/kg dose group, but had no effect on WT females in any dose group. Genotype alone influenced ultrasonic vocalizations, self-grooming and to a lesser extent sociability. Genotype alone also influenced effects of PCBs on sociability. PCB levels in the brain tissue of pups increased in a dose-dependent manner, but within any dose group did not differ between genotypes. In summary, developmental PCB exposure phenocopied social behavior phenotypes observed in mice expressing human mutations that modify intracellular Ca2+ dynamics, and expression of these mutations alleviated PCB effects on ultrasonic vocalizations and repetitive behavior, and modified the dose-response relationships and sex-dependent effects of PCB effects on social behavior. These findings suggest that: (1) developmental PCB exposure causes behavioral phenotypes that vary by sex and genotype; and (2) sex-specific responses to environmental factors may contribute to sex biases in the prevalence and/or severity of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Sunjay Sethi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Kimberly P. Keil Stietz
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Anthony E. Valenzuela
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Carolyn R. Klocke
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Jill L. Silverman
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
- The MIND Institute, University of California, Davis, Davis, CA, United States
| | - Birgit Puschner
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Isaac N. Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- The MIND Institute, University of California, Davis, Davis, CA, United States
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- The MIND Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
227
|
Li Y, Li S, Liu J, Huo Y, Luo XJ. The schizophrenia susceptibility gene NAGA regulates dendritic spine density: further evidence for the dendritic spine pathology of schizophrenia. Mol Psychiatry 2021; 26:7102-7104. [PMID: 34376824 DOI: 10.1038/s41380-021-01261-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Yifan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shiwu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yongxia Huo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, China. .,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
228
|
Wang DO. Epitranscriptomic regulation of cognitive development and decline. Semin Cell Dev Biol 2021; 129:3-13. [PMID: 34857470 DOI: 10.1016/j.semcdb.2021.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 11/24/2022]
Abstract
Functional genomics and systems biology have opened new doors to previously inaccessible genomic information and holistic approaches to study complex networks of genes and proteins in the central nervous system. The advances are revolutionizing our understanding of the genetic underpinning of cognitive development and decline by facilitating identifications of novel molecular regulators and physiological pathways underlying brain function, and by associating polymorphism and mutations to cognitive dysfunction and neurological diseases. However, our current understanding of these complex gene regulatory mechanisms has yet lacked sufficient mechanistic resolution for further translational breakthroughs. Here we review recent findings from the burgeoning field of epitranscriptomics in association of cognitive functions with a special focus on the epitranscritomic regulation in subcellular locations such as chromosome, synapse, and mitochondria. Although there are important gaps in knowledge, current evidence is suggesting that this layer of RNA regulation may be of particular interest for the spatiotemporally coordinated regulation of gene networks in developing and maintaining brain function that underlie cognitive changes.
Collapse
Affiliation(s)
- Dan Ohtan Wang
- Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Graduate School of Biostudies, Kyoto University, Yoshida Hon-machi, Kyoto 606-8501, Japan.
| |
Collapse
|
229
|
Significance of GABA A Receptor for Cognitive Function and Hippocampal Pathology. Int J Mol Sci 2021; 22:ijms222212456. [PMID: 34830337 PMCID: PMC8623595 DOI: 10.3390/ijms222212456] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 02/05/2023] Open
Abstract
The hippocampus is a primary area for contextual memory, known to process spatiotemporal information within a specific episode. Long-term strengthening of glutamatergic transmission is a mechanism of contextual learning in the dorsal cornu ammonis 1 (CA1) area of the hippocampus. CA1-specific immobilization or blockade of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptor delivery can impair learning performance, indicating a causal relationship between learning and receptor delivery into the synapse. Moreover, contextual learning also strengthens GABAA (gamma-aminobutyric acid) receptor-mediated inhibitory synapses onto CA1 neurons. Recently we revealed that strengthening of GABAA receptor-mediated inhibitory synapses preceded excitatory synaptic plasticity after contextual learning, resulting in a reduced synaptic excitatory/inhibitory (E/I) input balance that returned to pretraining levels within 10 min. The faster plasticity at inhibitory synapses may allow encoding a contextual memory and prevent cognitive dysfunction in various hippocampal pathologies. In this review, we focus on the dynamic changes of GABAA receptor mediated-synaptic currents after contextual learning and the intracellular mechanism underlying rapid inhibitory synaptic plasticity. In addition, we discuss that several pathologies, such as Alzheimer’s disease, autism spectrum disorders and epilepsy are characterized by alterations in GABAA receptor trafficking, synaptic E/I imbalance and neuronal excitability.
Collapse
|
230
|
Mirabella F, Desiato G, Mancinelli S, Fossati G, Rasile M, Morini R, Markicevic M, Grimm C, Amegandjin C, Termanini A, Peano C, Kunderfranco P, di Cristo G, Zerbi V, Menna E, Lodato S, Matteoli M, Pozzi D. Prenatal interleukin 6 elevation increases glutamatergic synapse density and disrupts hippocampal connectivity in offspring. Immunity 2021; 54:2611-2631.e8. [PMID: 34758338 PMCID: PMC8585508 DOI: 10.1016/j.immuni.2021.10.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/24/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023]
Abstract
Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.
Collapse
Affiliation(s)
- Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Genni Desiato
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Sara Mancinelli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giuliana Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Raffaella Morini
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marija Markicevic
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Christina Grimm
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Clara Amegandjin
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alberto Termanini
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Clelia Peano
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, 20089 Rozzano, Milan, Italy; Genomic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Graziella di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Valerio Zerbi
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland; Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich 8057, Switzerland
| | - Elisabetta Menna
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy.
| | - Davide Pozzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
231
|
Zhang YQ, Lin WP, Huang LP, Zhao B, Zhang CC, Yin DM. Dopamine D2 receptor regulates cortical synaptic pruning in rodents. Nat Commun 2021; 12:6444. [PMID: 34750364 PMCID: PMC8576001 DOI: 10.1038/s41467-021-26769-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 10/22/2021] [Indexed: 12/22/2022] Open
Abstract
Synaptic pruning during adolescence is important for appropriate neurodevelopment and synaptic plasticity. Aberrant synaptic pruning may underlie a variety of brain disorders such as schizophrenia, autism and anxiety. Dopamine D2 receptor (Drd2) is associated with several neuropsychiatric diseases and is the target of some antipsychotic drugs. Here we generate self-reporting Drd2 heterozygous (SR-Drd2+/-) rats to simultaneously visualize Drd2-positive neurons and downregulate Drd2 expression. Time course studies on the developing anterior cingulate cortex (ACC) from control and SR-Drd2+/- rats reveal important roles of Drd2 in regulating synaptic pruning rather than synapse formation. Drd2 also regulates LTD, a form of synaptic plasticity which includes some similar cellular/biochemical processes as synaptic pruning. We further demonstrate that Drd2 regulates synaptic pruning via cell-autonomous mechanisms involving activation of mTOR signaling. Deficits of Drd2-mediated synaptic pruning in the ACC during adolescence lead to hyper-glutamatergic function and anxiety-like behaviors in adulthood. Taken together, our results demonstrate important roles of Drd2 in cortical synaptic pruning.
Collapse
Affiliation(s)
- Ya-Qiang Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China
| | - Wei-Peng Lin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China
- Joint Translational Science and Technology Research Institute, East China Normal University, 200062, Shanghai, China
| | - Li-Ping Huang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China
| | - Bing Zhao
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China
| | - Cheng-Cheng Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China
| | - Dong-Min Yin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, 200062, Shanghai, China.
| |
Collapse
|
232
|
Verma V, Kumar MJV, Sharma K, Rajaram S, Muddashetty R, Manjithaya R, Behnisch T, Clement JP. Pharmacological intervention in young adolescents rescues synaptic physiology and behavioural deficits in Syngap1 +/- mice. Exp Brain Res 2021; 240:289-309. [PMID: 34739555 DOI: 10.1007/s00221-021-06254-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/21/2021] [Indexed: 01/04/2023]
Abstract
Haploinsufficiency in SYNGAP1 is implicated in intellectual disability (ID) and autism spectrum disorder (ASD) and affects the maturation of dendritic spines. The abnormal spine development has been suggested to cause a disbalance of excitatory and inhibitory (E/I) neurotransmission at distinct developmental periods. In addition, E/I imbalances in Syngap1+/- mice might be due to abnormalities in K+-Cl- co-transporter function (NKCC1, KCC2), in a maner similar to the murine models of Fragile-X and Rett syndromes. To study whether an altered intracellular chloride ion concentration represents an underlying mechanism of modified function of GABAergic synapses in Dentate Gyrus Granule Cells of Syngap1+/- recordings were performed at different developmental stages of the mice. We observed depolarised neurons at P14-15 as illustrated by decreased Cl- reversal potential in Syngap1+/- mice. The KCC2 expression was decreased compared to Wild-type (WT) mice at P14-15. The GSK-3β inhibitor, 6-bromoindirubin-3'-oxime (6BIO) that crosses the blood-brain barrier, was tested to restore the function of GABAergic synapses. We discovered that the intraperitoneal administration of 6BIO during the critical period or young adolescents [P30 to P80 (4-week to 10-week)] normalised an altered E/I balance, the deficits of synaptic plasticity, and behavioural performance like social novelty, anxiety, and memory of the Syngap1+/- mice. In summary, altered GABAergic function in Syngap1+/- mice is due to reduced KCC2 expression leading to an increase in the intracellular chloride concentration that can be counteracted by the 6BIO, which restored cognitive, emotional, and social symptoms by pharmacological intervention, particularly in adulthood.
Collapse
Affiliation(s)
- Vijaya Verma
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - M J Vijay Kumar
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Kavita Sharma
- International Centre for Material Sciences, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Sridhar Rajaram
- International Centre for Material Sciences, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Ravi Muddashetty
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India
| | - Ravi Manjithaya
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India.,Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Thomas Behnisch
- Institutes of Brain Sciences, Fudan University, Shanghai, 200032, China
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India.
| |
Collapse
|
233
|
Soteros BM, Sia GM. Complement and microglia dependent synapse elimination in brain development. WIREs Mech Dis 2021; 14:e1545. [PMID: 34738335 PMCID: PMC9066608 DOI: 10.1002/wsbm.1545] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 01/31/2023]
Abstract
Synapse elimination, also known as synaptic pruning, is a critical step in the maturation of neural circuits during brain development. Mounting evidence indicates that the complement cascade of the innate immune system plays an important role in synapse elimination. Studies indicate that excess synapses during development are opsonized by complement proteins and subsequently phagocytosed by microglia which expresses complement receptors. The process is regulated by diverse molecular signals, including complement inhibitors that affect the activation of complement, as well as signals that affect microglial recruitment and activation. These signals may promote or inhibit the removal of specific sets of synapses during development. The complement-microglia system has also been implicated in the pathogenesis of several developmental brain disorders, suggesting that the dysregulation of mechanisms of synapse pruning may underlie the specific circuitry defects in these diseases. Here, we review the latest evidence on the molecular and cellular mechanisms of complement-dependent and microglia-dependent synapse elimination during brain development, and highlight the potential of this system as a therapeutic target for developmental brain disorders. This article is categorized under: Neurological Diseases > Molecular and Cellular Physiology Neurological Diseases > Stem Cells and Development Immune System Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Breeanne M Soteros
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Gek Ming Sia
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
234
|
Biane C, Rückerl F, Abrahamsson T, Saint-Cloment C, Mariani J, Shigemoto R, DiGregorio DA, Sherrard RM, Cathala L. Developmental emergence of two-stage nonlinear synaptic integration in cerebellar interneurons. eLife 2021; 10:65954. [PMID: 34730085 PMCID: PMC8565927 DOI: 10.7554/elife.65954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Synaptic transmission, connectivity, and dendritic morphology mature in parallel during brain development and are often disrupted in neurodevelopmental disorders. Yet how these changes influence the neuronal computations necessary for normal brain function are not well understood. To identify cellular mechanisms underlying the maturation of synaptic integration in interneurons, we combined patch-clamp recordings of excitatory inputs in mouse cerebellar stellate cells (SCs), three-dimensional reconstruction of SC morphology with excitatory synapse location, and biophysical modeling. We found that postnatal maturation of postsynaptic strength was homogeneously reduced along the somatodendritic axis, but dendritic integration was always sublinear. However, dendritic branching increased without changes in synapse density, leading to a substantial gain in distal inputs. Thus, changes in synapse distribution, rather than dendrite cable properties, are the dominant mechanism underlying the maturation of neuronal computation. These mechanisms favor the emergence of a spatially compartmentalized two-stage integration model promoting location-dependent integration within dendritic subunits.
Collapse
Affiliation(s)
- Celia Biane
- Sorbonne Université et CNRS UMR 8256, Adaptation Biologique et Vieillissement, Paris, France
| | - Florian Rückerl
- Institut Pasteur, Université de Paris, CNRS UMR 3571, Unit of Synapse and Circuit Dynamics, Paris, France
| | - Therese Abrahamsson
- Institut Pasteur, Université de Paris, CNRS UMR 3571, Unit of Synapse and Circuit Dynamics, Paris, France
| | - Cécile Saint-Cloment
- Institut Pasteur, Université de Paris, CNRS UMR 3571, Unit of Synapse and Circuit Dynamics, Paris, France
| | - Jean Mariani
- Sorbonne Université et CNRS UMR 8256, Adaptation Biologique et Vieillissement, Paris, France
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - David A DiGregorio
- Institut Pasteur, Université de Paris, CNRS UMR 3571, Unit of Synapse and Circuit Dynamics, Paris, France
| | - Rachel M Sherrard
- Sorbonne Université et CNRS UMR 8256, Adaptation Biologique et Vieillissement, Paris, France
| | - Laurence Cathala
- Sorbonne Université et CNRS UMR 8256, Adaptation Biologique et Vieillissement, Paris, France.,Paris Brain Institute, CNRS UMR 7225 - Inserm U1127 - Sorbonne Université Groupe Hospitalier Pitié Salpêtrière, Paris, France
| |
Collapse
|
235
|
Li X, Zhong H, Wang Z, Xiao R, Antonson P, Liu T, Wu C, Zou J, Wang L, Nalvarte I, Xu H, Warner M, Gustafsson JA, Fan X. Loss of liver X receptor β in astrocytes leads to anxiety-like behaviors via regulating synaptic transmission in the medial prefrontal cortex in mice. Mol Psychiatry 2021; 26:6380-6393. [PMID: 33963286 DOI: 10.1038/s41380-021-01139-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 04/08/2021] [Accepted: 04/19/2021] [Indexed: 02/03/2023]
Abstract
Astrocytes are integral components of synaptic transmission, and their dysfunction leads to neuropsychiatric disorders such as anxiety and depression. Liver X receptor β (LXRβ) is expressed in astrocytes, and LXRβ global knockout mice shows impaired synaptic formation. In order to define the role of LXRβ in astrocytes, we used a conditional Cre-loxP system to specifically remove LXRβ from astrocytes. We found that this deletion caused anxiety-like but not depressive-like behaviors in adult male mice. This behavioral phenotype could be completely reproduced by selective deletion of LXRβ in astrocytes in the medial prefrontal cortex (mPFC). Pyramidal neurons in layer V of mPFC are involved in mood behaviors. We found that there was an increased spontaneous excitatory synaptic transmission in layer V pyramidal neurons of the mPFC of these mice. This was concurrent with increased dendritic complexity, despite normal appearance and number of dendritic spines. In addition, gene ontology analysis of RNA sequencing revealed that deletion of astrocytic LXRβ led to the enrichment of the process of synaptic transmission in mPFC. Finally, we also confirmed that renormalized excitatory synaptic transmission in layer V pyramidal neurons alleviated the anxiety in mice with astrocytic LXRβ deletion in mPFC. Together, our findings reveal that astrocytic LXRβ in mPFC is critical in the regulation of synaptic transmission, and this provides a potential new target for treatment of anxiety-like behavior.
Collapse
Affiliation(s)
- Xin Li
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Hongyu Zhong
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Zhongke Wang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Rui Xiao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Per Antonson
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Tianyao Liu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Chuan Wu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Jiao Zou
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Lian Wang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Ivan Nalvarte
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Margaret Warner
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Jan-Ake Gustafsson
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden. .,Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, PR China.
| |
Collapse
|
236
|
Wang JS, Kamath T, Mazur CM, Mirzamohammadi F, Rotter D, Hojo H, Castro CD, Tokavanich N, Patel R, Govea N, Enishi T, Wu Y, da Silva Martins J, Bruce M, Brooks DJ, Bouxsein ML, Tokarz D, Lin CP, Abdul A, Macosko EZ, Fiscaletti M, Munns CF, Ryder P, Kost-Alimova M, Byrne P, Cimini B, Fujiwara M, Kronenberg HM, Wein MN. Control of osteocyte dendrite formation by Sp7 and its target gene osteocrin. Nat Commun 2021; 12:6271. [PMID: 34725346 PMCID: PMC8560803 DOI: 10.1038/s41467-021-26571-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 10/12/2021] [Indexed: 02/05/2023] Open
Abstract
Some osteoblasts embed within bone matrix, change shape, and become dendrite-bearing osteocytes. The circuitry that drives dendrite formation during "osteocytogenesis" is poorly understood. Here we show that deletion of Sp7 in osteoblasts and osteocytes causes defects in osteocyte dendrites. Profiling of Sp7 target genes and binding sites reveals unexpected repurposing of this transcription factor to drive dendrite formation. Osteocrin is a Sp7 target gene that promotes osteocyte dendrite formation and rescues defects in Sp7-deficient mice. Single-cell RNA-sequencing demonstrates defects in osteocyte maturation in the absence of Sp7. Sp7-dependent osteocyte gene networks are associated with human skeletal diseases. Moreover, humans with a SP7R316C mutation show defective osteocyte morphology. Sp7-dependent genes that mark osteocytes are enriched in neurons, highlighting shared features between osteocytic and neuronal connectivity. These findings reveal a role for Sp7 and its target gene Osteocrin in osteocytogenesis, revealing that pathways that control osteocyte development influence human bone diseases.
Collapse
Affiliation(s)
- Jialiang S Wang
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tushar Kamath
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Courtney M Mazur
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fatemeh Mirzamohammadi
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Plastic and Reconstructive Surgery, Wright State University, Dayton, OH, USA
| | - Daniel Rotter
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- University of Applied Sciences Technikum Wien, Vienna, Austria
| | - Hironori Hojo
- Center for Disease Biology and Integrative Medicine, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Christian D Castro
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicha Tokavanich
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rushi Patel
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicolas Govea
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY, USA
| | - Tetsuya Enishi
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Orthopedic Surgery, Tokushima Municipal Hospital, Tokushima, Japan
| | - Yunshu Wu
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | | | - Michael Bruce
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel J Brooks
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Advanced Orthopedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MaA, USA
| | - Mary L Bouxsein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Advanced Orthopedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MaA, USA
| | - Danielle Tokarz
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, Saint Mary's University, Halifax, Canada
| | - Charles P Lin
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Abdul Abdul
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Evan Z Macosko
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Melissa Fiscaletti
- Pediatric Department, Sainte-Justine University Hospital Centre, Montreal, Canada
| | - Craig F Munns
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, NSW, Australia
- Discipline of Paediatrics & Child Health, University of Sydney, Sydney, 2006, Australia
| | - Pearl Ryder
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Imaging Platform, Cambridge, MA, USA
| | - Maria Kost-Alimova
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Center for the Development of Therapeutics, Cambridge, MA, USA
| | - Patrick Byrne
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Center for the Development of Therapeutics, Cambridge, MA, USA
| | - Beth Cimini
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Imaging Platform, Cambridge, MA, USA
| | - Makoto Fujiwara
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Henry M Kronenberg
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
237
|
Huang SY, Witzel T, Keil B, Scholz A, Davids M, Dietz P, Rummert E, Ramb R, Kirsch JE, Yendiki A, Fan Q, Tian Q, Ramos-Llordén G, Lee HH, Nummenmaa A, Bilgic B, Setsompop K, Wang F, Avram AV, Komlosh M, Benjamini D, Magdoom KN, Pathak S, Schneider W, Novikov DS, Fieremans E, Tounekti S, Mekkaoui C, Augustinack J, Berger D, Shapson-Coe A, Lichtman J, Basser PJ, Wald LL, Rosen BR. Connectome 2.0: Developing the next-generation ultra-high gradient strength human MRI scanner for bridging studies of the micro-, meso- and macro-connectome. Neuroimage 2021; 243:118530. [PMID: 34464739 PMCID: PMC8863543 DOI: 10.1016/j.neuroimage.2021.118530] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/10/2021] [Accepted: 08/27/2021] [Indexed: 11/26/2022] Open
Abstract
The first phase of the Human Connectome Project pioneered advances in MRI technology for mapping the macroscopic structural connections of the living human brain through the engineering of a whole-body human MRI scanner equipped with maximum gradient strength of 300 mT/m, the highest ever achieved for human imaging. While this instrument has made important contributions to the understanding of macroscale connectional topology, it has also demonstrated the potential of dedicated high-gradient performance scanners to provide unparalleled in vivo assessment of neural tissue microstructure. Building on the initial groundwork laid by the original Connectome scanner, we have now embarked on an international, multi-site effort to build the next-generation human 3T Connectome scanner (Connectome 2.0) optimized for the study of neural tissue microstructure and connectional anatomy across multiple length scales. In order to maximize the resolution of this in vivo microscope for studies of the living human brain, we will push the diffusion resolution limit to unprecedented levels by (1) nearly doubling the current maximum gradient strength from 300 mT/m to 500 mT/m and tripling the maximum slew rate from 200 T/m/s to 600 T/m/s through the design of a one-of-a-kind head gradient coil optimized to minimize peripheral nerve stimulation; (2) developing high-sensitivity multi-channel radiofrequency receive coils for in vivo and ex vivo human brain imaging; (3) incorporating dynamic field monitoring to minimize image distortions and artifacts; (4) developing new pulse sequences to integrate the strongest diffusion encoding and highest spatial resolution ever achieved in the living human brain; and (5) calibrating the measurements obtained from this next-generation instrument through systematic validation of diffusion microstructural metrics in high-fidelity phantoms and ex vivo brain tissue at progressively finer scales with accompanying diffusion simulations in histology-based micro-geometries. We envision creating the ultimate diffusion MRI instrument capable of capturing the complex multi-scale organization of the living human brain - from the microscopic scale needed to probe cellular geometry, heterogeneity and plasticity, to the mesoscopic scale for quantifying the distinctions in cortical structure and connectivity that define cyto- and myeloarchitectonic boundaries, to improvements in estimates of macroscopic connectivity.
Collapse
Affiliation(s)
- Susie Y Huang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | | | - Boris Keil
- Institute of Medical Physics and Radiation Protection (IMPS), TH-Mittelhessen University of Applied Sciences (THM), Giessen, Germany
| | - Alina Scholz
- Institute of Medical Physics and Radiation Protection (IMPS), TH-Mittelhessen University of Applied Sciences (THM), Giessen, Germany
| | - Mathias Davids
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - John E Kirsch
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anastasia Yendiki
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Qiuyun Fan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Qiyuan Tian
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gabriel Ramos-Llordén
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hong-Hsi Lee
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aapo Nummenmaa
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Berkin Bilgic
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kawin Setsompop
- Radiological Sciences Laboratory, Department of Radiology, Stanford University, Stanford, CA, USA
| | - Fuyixue Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexandru V Avram
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Michal Komlosh
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Dan Benjamini
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Kulam Najmudeen Magdoom
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Sudhir Pathak
- Learning Research and Development Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Walter Schneider
- Learning Research and Development Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dmitry S Novikov
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI2R), New York University School of Medicine, New York, NY, USA
| | - Els Fieremans
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI2R), New York University School of Medicine, New York, NY, USA
| | - Slimane Tounekti
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Choukri Mekkaoui
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jean Augustinack
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel Berger
- Department of Molecular and Cell Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Alexander Shapson-Coe
- Department of Molecular and Cell Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Jeff Lichtman
- Department of Molecular and Cell Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Peter J Basser
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Lawrence L Wald
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
238
|
Faust TE, Gunner G, Schafer DP. Mechanisms governing activity-dependent synaptic pruning in the developing mammalian CNS. Nat Rev Neurosci 2021; 22:657-673. [PMID: 34545240 PMCID: PMC8541743 DOI: 10.1038/s41583-021-00507-y] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Almost 60 years have passed since the initial discovery by Hubel and Wiesel that changes in neuronal activity can elicit developmental rewiring of the central nervous system (CNS). Over this period, we have gained a more comprehensive picture of how both spontaneous neural activity and sensory experience-induced changes in neuronal activity guide CNS circuit development. Here we review activity-dependent synaptic pruning in the mammalian CNS, which we define as the removal of a subset of synapses, while others are maintained, in response to changes in neural activity in the developing nervous system. We discuss the mounting evidence that immune and cell-death molecules are important mechanistic links by which changes in neural activity guide the pruning of specific synapses, emphasizing the role of glial cells in this process. Finally, we discuss how these developmental pruning programmes may go awry in neurodevelopmental disorders of the human CNS, focusing on autism spectrum disorder and schizophrenia. Together, our aim is to give an overview of how the field of activity-dependent pruning research has evolved, led to exciting new questions and guided the identification of new, therapeutically relevant mechanisms that result in aberrant circuit development in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Travis E Faust
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Georgia Gunner
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
239
|
Moniem Ali R, El-Wakeel HA, Al-Saleh DF, Shukri MI, Ansari KMN. Autism spectrum disorder in architecture perspective: a review of the literature and bibliometric assessment of research indexed in Web of Science. F1000Res 2021; 10:1087. [PMID: 35923662 PMCID: PMC9326241 DOI: 10.12688/f1000research.54437.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/09/2021] [Indexed: 09/02/2024] Open
Abstract
Purpose: The primary objective of this research paper was to explore the current state-of-the-art research on autism spectrum disorder from a designer's perspective. An increasing number of scholarly publications in this discipline have urged researcher interest in this topic; however, there is still a lack of quantitative analysis. Therefore, this paper aims to analyze global research output on autism spectrum disorder from a designer's perspective during 1992-2021. Methodology: A bibliometric method was employed to analyze the published literature from 1992-2021. 812 papers were downloaded from the Web of Science core collection for analysis focused on annual growth of literature, prolific authors, authorship pattern, productive organizations, countries, international collaboration, literature trends by keyword analysis, and identifying the funding agencies. Various bibliometrics and scientometrics software were used to analyze the data, namely Bibexcel, Biblioshiny, and VOS viewer. Results: There were 812 research papers published in 405 sources during 1992-2021. 2019 was noted as the most productive year (NP=101), and 2014 received the highest number of citations (TC=6634). Researchers preferred to publish as journal articles (NP=538; TC=24922). The University of Toronto, Canada, was identified as a productive institution with 42 publications and 5358 citations. The USA was the leading producing country with 433 publications, and most of the researchers publish their work in the journal "Scientific Reports" (NP=16). The word "autism" (NP=257) and "architecture" (NP=165) were the most frequently used keywords in autism research.
Collapse
Affiliation(s)
- Reham Moniem Ali
- Interior Design Department, College of Design, Imam Abdulrahman bin Faisal University, Saudi Arabia, Eastern Province, PO. 1982, Saudi Arabia
| | - Hala A. El-Wakeel
- Interior Design Department, College of Design, Imam Abdulrahman bin Faisal University, Saudi Arabia, Eastern Province, PO. 1982, Saudi Arabia
| | - Deema Faisal Al-Saleh
- Interior Design Department, College of Design, Imam Abdulrahman bin Faisal University, Saudi Arabia, Eastern Province, PO. 1982, Saudi Arabia
| | - Mai Ibrahim Shukri
- Interior Design Department, College of Design, Imam Abdulrahman bin Faisal University, Saudi Arabia, Eastern Province, PO. 1982, Saudi Arabia
| | - Khadeeja M N Ansari
- Interior Design Department, College of Design, Imam Abdulrahman bin Faisal University, Saudi Arabia, Eastern Province, PO. 1982, Saudi Arabia
| |
Collapse
|
240
|
Moniem Ali R, El-Wakeel HA, Al-Saleh DF, Shukri MI, Ansari KMN. Autism spectrum disorder in architecture perspective: a review of the literature and bibliometric assessment of research indexed in Web of Science. F1000Res 2021; 10:1087. [PMID: 35923662 PMCID: PMC9326241 DOI: 10.12688/f1000research.54437.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2022] [Indexed: 11/20/2022] Open
Abstract
PURPOSE An increasing number of scholarly publications on autism spectrum disorder (ASD) have urged researcher interest in this topic; however, there is still a lack of quantitative analysis. Therefore, this study aims to cover the knowledge gap between the amount of literature published on ASD research on architectural and designers' perspectives compared to the medical and psychological fields. The study has analyzed global research output on ASD from a designer's perspective to recognize this gap related to designing the physical environment. Methodology: The bibliometric method was employed to analyze the published literature from 1992-to 2021. 812 papers were downloaded from the Web of Science for analysis based on annual growth of literature, prolific authors, authorship pattern, organizations, countries, international collaboration, and subject development by keywords and thematic map analyses. Various bibliometric and scientometric software was used to analyze the data, namely Bibexcel, Biblioshiny, and VOS viewer. RESULTS The812 research papers were published in 405 sources. 2019 appeared as a productive year (NP=101), and 2014 received the highest number of citations (TC=6634). Researchers preferred to publish as journal articles (NP=538; TC=24922). The University of Toronto, Canada, was identified as a productive institution with 42 publications and 5358 citations. The USA was the leading producing country with 433 publications, and most of the researchers published in the journal " Scientific Reports" (NP=16). The word autism (NP=257) and architecture (NP=165) were more frequently used keywords. CONCLUSION The study identified a massive gap in the development of literature in ASD for architecture design and built environment perspective, the most important and trending keywords are missing, and the analyses also showed a lack of subject development. The authors have suggested areas and keywords for further research to fulfill the gap in the future.
Collapse
Affiliation(s)
- Reham Moniem Ali
- Interior Design Department, College of Design, Imam Abdulrahman bin Faisal University, Saudi Arabia, Eastern Province, PO. 1982, Saudi Arabia
| | - Hala A. El-Wakeel
- Interior Design Department, College of Design, Imam Abdulrahman bin Faisal University, Saudi Arabia, Eastern Province, PO. 1982, Saudi Arabia
| | - Deema Faisal Al-Saleh
- Interior Design Department, College of Design, Imam Abdulrahman bin Faisal University, Saudi Arabia, Eastern Province, PO. 1982, Saudi Arabia
| | - Mai Ibrahim Shukri
- Interior Design Department, College of Design, Imam Abdulrahman bin Faisal University, Saudi Arabia, Eastern Province, PO. 1982, Saudi Arabia
| | - Khadeeja M N Ansari
- Interior Design Department, College of Design, Imam Abdulrahman bin Faisal University, Saudi Arabia, Eastern Province, PO. 1982, Saudi Arabia
| |
Collapse
|
241
|
Vallés AS, Barrantes FJ. Dendritic spine membrane proteome and its alterations in autistic spectrum disorder. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:435-474. [PMID: 35034726 DOI: 10.1016/bs.apcsb.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dendritic spines are small protrusions stemming from the dendritic shaft that constitute the primary specialization for receiving and processing excitatory neurotransmission in brain synapses. The disruption of dendritic spine function in several neurological and neuropsychiatric diseases leads to severe information-processing deficits with impairments in neuronal connectivity and plasticity. Spine dysregulation is usually accompanied by morphological alterations to spine shape, size and/or number that may occur at early pathophysiological stages and not necessarily be reflected in clinical manifestations. Autism spectrum disorder (ASD) is one such group of diseases involving changes in neuronal connectivity and abnormal morphology of dendritic spines on postsynaptic neurons. These alterations at the subcellular level correlate with molecular changes in the spine proteome, with alterations in the copy number, topography, or in severe cases in the phenotype of the molecular components, predominantly of those proteins involved in spine recognition and adhesion, reflected in abnormally short lifetimes of the synapse and compensatory increases in synaptic connections. Since cholinergic neurotransmission participates in the regulation of cognitive function (attention, memory, learning processes, cognitive flexibility, social interactions) brain acetylcholine receptors are likely to play an important role in the dysfunctional synapses in ASD, either directly or indirectly via the modulatory functions exerted on other neurotransmitter receptor proteins and spine-resident proteins.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), Bahía Blanca, Argentina
| | - Francisco J Barrantes
- Instituto de Investigaciones Biomédicas (BIOMED), UCA-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
242
|
Aswendt M, Green C, Sadler R, Llovera G, Dzikowski L, Heindl S, Gomez de Agüero M, Diedenhofen M, Vogel S, Wieters F, Wiedermann D, Liesz A, Hoehn M. The gut microbiota modulates brain network connectivity under physiological conditions and after acute brain ischemia. iScience 2021; 24:103095. [PMID: 34622150 PMCID: PMC8479691 DOI: 10.1016/j.isci.2021.103095] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/14/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
The gut microbiome has been implicated as a key regulator of brain function in health and disease. But the impact of gut microbiota on functional brain connectivity is unknown. We used resting-state functional magnetic resonance imaging in germ-free and normally colonized mice under naive conditions and after ischemic stroke. We observed a strong, brain-wide increase of functional connectivity in germ-free animals. Graph theoretical analysis revealed significant higher values in germ-free animals, indicating a stronger and denser global network but with less structural organization. Breakdown of network function after stroke equally affected germ-free and colonized mice. Results from histological analyses showed changes in dendritic spine densities, as well as an immature microglial phenotype, indicating impaired microglia-neuron interaction in germ-free mice as potential cause of this phenomenon. These results demonstrate the substantial impact of bacterial colonization on brain-wide function and extend our so far mainly (sub) cellular understanding of the gut-brain axis.
Collapse
Affiliation(s)
- Markus Aswendt
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital, 50923 Cologne, Germany
| | - Claudia Green
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Rebecca Sadler
- Institute for Stroke and Dementia Research (ISD), LMU Munich, Feodor-Lynen Strasse 17, 81377 Munich, Germany
| | - Gemma Llovera
- Institute for Stroke and Dementia Research (ISD), LMU Munich, Feodor-Lynen Strasse 17, 81377 Munich, Germany
| | - Lauren Dzikowski
- Institute for Stroke and Dementia Research (ISD), LMU Munich, Feodor-Lynen Strasse 17, 81377 Munich, Germany
| | - Steffanie Heindl
- Institute for Stroke and Dementia Research (ISD), LMU Munich, Feodor-Lynen Strasse 17, 81377 Munich, Germany
| | - Mercedes Gomez de Agüero
- Department for BioMedical Research (DBMR), University of Bern, 3012 Bern, Switzerland
- Institute of Systems Immunology, Julius-Maximilians University of Würzburg, 97070 Würzburg, Germany
| | - Michael Diedenhofen
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Stefanie Vogel
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Frederique Wieters
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital, 50923 Cologne, Germany
| | - Dirk Wiedermann
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), LMU Munich, Feodor-Lynen Strasse 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 80807 Munich, Germany
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| |
Collapse
|
243
|
ERK/MAPK signalling in the developing brain: Perturbations and consequences. Neurosci Biobehav Rev 2021; 131:792-805. [PMID: 34634357 DOI: 10.1016/j.neubiorev.2021.10.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/26/2021] [Accepted: 10/05/2021] [Indexed: 12/18/2022]
Abstract
The extracellular regulated kinase/microtubule-associated protein kinase (ERK/MAPK) signalling pathway transduces signals that cause an alteration in the ongoing metabolic pathways and modifies gene expression patterns; thus, influencing cellular behaviour. ERK/MAPK signalling is essential for the proper development of the nervous system from neural progenitor cells derived from the embryonic mesoderm. Several signalling molecules that regulate the well-coordinated process of neurodevelopment transduce developmental information through the ERK/MAPK signalling pathway. The ERK/MAPK is a potential novel therapeutic target in several neurodevelopmental disorders, however, despite years of study, there is still significant uncertainty about the exact mechanism by which the ERK/MAPK signalling pathway elicits specific responses in neurodevelopment. Here, we will review the evidence highlighting the role of ERK/MAPK signalling in neurodevelopment. We will also discuss the structural implication and behavioural deficits associated with perturbed ERK/MAPK signalling pathway in cortical development, whilst examining its contribution to the neuropathology of several neurodevelopmental disorders, such as Autism Spectrum Disorder, Schizophrenia, Fragile X, and Attention Deficit Hyperactive Disorder.
Collapse
|
244
|
Barón-Mendoza I, Maqueda-Martínez E, Martínez-Marcial M, De la Fuente-Granada M, Gómez-Chavarin M, González-Arenas A. Changes in the Number and Morphology of Dendritic Spines in the Hippocampus and Prefrontal Cortex of the C58/J Mouse Model of Autism. Front Cell Neurosci 2021; 15:726501. [PMID: 34616277 PMCID: PMC8488392 DOI: 10.3389/fncel.2021.726501] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/25/2021] [Indexed: 11/22/2022] Open
Abstract
Autism spectrum disorder (ASD) has a broad range of neurobiological characteristics, including alterations in dendritic spines, where approximately 90% of excitatory synapses occur. Therefore, changes in their number or morphology would be related to atypical brain communication. The C58/J inbred mouse strain displays low sociability, impaired communication, and stereotyped behavior; hence, it is considered among the animal models suitable for the study of idiopathic autism. Thus, this study aimed to evaluate the dendritic spine differences in the hippocampus and the prefrontal cortex of C58/J mice. We found changes in the number of spines and morphology in a brain region-dependent manner: a subtle decrease in spine density in the prefrontal cortex, higher frequency of immature phenotype spines characterized by filopodia-like length or small morphology, and a lower number of mature phenotype spines with mushroom-like or wide heads in the hippocampus. Moreover, an in silico analysis showed single nucleotide polymorphisms (SNPs) at genes collectively involved in regulating structural plasticity with a likely association with ASD, including MAP1A (Microtubule-Associated Protein 1A), GRM7 (Metabotropic Glutamate Receptor, 7), ANKRD11 (Ankyrin Repeat Domain 11), and SLC6A4 (Solute Carrier Family 6, member 4), which might support the relationship between the C58/J strain genome, an autistic-like behavior, and the observed anomalies in the dendritic spines.
Collapse
Affiliation(s)
- Isabel Barón-Mendoza
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Emely Maqueda-Martínez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mónica Martínez-Marcial
- Unidad de Modelos Biológicos, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marisol De la Fuente-Granada
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Margarita Gómez-Chavarin
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Aliesha González-Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
245
|
Lee LC, Su MT, Huang HY, Cho YC, Yeh TK, Chang CY. Association of CaMK2A and MeCP2 signaling pathways with cognitive ability in adolescents. Mol Brain 2021; 14:152. [PMID: 34607601 PMCID: PMC8491411 DOI: 10.1186/s13041-021-00858-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/13/2021] [Indexed: 11/28/2022] Open
Abstract
The glutamatergic signaling pathway is involved in molecular learning and human cognitive ability. Specific single variants (SNVs, formerly single-nucleotide polymorphisms) in the genes encoding N-methyl-d-aspartate receptor subunits have been associated with neuropsychiatric disorders by altering glutamate transmission. However, these variants associated with cognition and mental activity have rarely been explored in healthy adolescents. In this study, we screened for SNVs in the glutamatergic signaling pathway to identify genetic variants associated with cognitive ability. We found that SNVs in the subunits of ionotropic glutamate receptors, including GRIA1, GRIN1, GRIN2B, GRIN2C, GRIN3A, GRIN3B, and calcium/calmodulin-dependent protein kinase IIα (CaMK2A) are associated with cognitive function. Plasma CaMK2A level was correlated positively with the cognitive ability of Taiwanese senior high school students. We demonstrated that elevating CaMK2A increased its autophosphorylation at T286 and increased the expression of its downstream targets, including GluA1 and phosphor- GluA1 in vivo. Additionally, methyl-CpG binding protein 2 (MeCP2), a downstream target of CaMK2A, was found to activate the expression of CaMK2A, suggesting that MeCP2 and CaMK2A can form a positive feedback loop. In summary, two members of the glutamatergic signaling pathway, CaMK2A and MeCP2, are implicated in the cognitive ability of adolescents; thus, altering the expression of CaMK2A may affect cognitive ability in youth.
Collapse
Affiliation(s)
- Li-Ching Lee
- Science Education Center and Graduate Institute of Science Education, National Taiwan Normal University, No. 88, Sec. 4, Ting-Chou Rd., Taipei, 11677, Taiwan, Republic of China
| | - Ming-Tsan Su
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hsing-Ying Huang
- Science Education Center and Graduate Institute of Science Education, National Taiwan Normal University, No. 88, Sec. 4, Ting-Chou Rd., Taipei, 11677, Taiwan, Republic of China
| | - Ying-Chun Cho
- Science Education Center and Graduate Institute of Science Education, National Taiwan Normal University, No. 88, Sec. 4, Ting-Chou Rd., Taipei, 11677, Taiwan, Republic of China
| | - Ting-Kuang Yeh
- Science Education Center and Graduate Institute of Science Education, National Taiwan Normal University, No. 88, Sec. 4, Ting-Chou Rd., Taipei, 11677, Taiwan, Republic of China. .,Institute of Marine Environment Science and Technology, National Taiwan Normal University, Taipei, Taiwan. .,Department of Earth Science, National Taiwan Normal University, Taipei, Taiwan.
| | - Chun-Yen Chang
- Science Education Center and Graduate Institute of Science Education, National Taiwan Normal University, No. 88, Sec. 4, Ting-Chou Rd., Taipei, 11677, Taiwan, Republic of China. .,Department of Earth Science, National Taiwan Normal University, Taipei, Taiwan.
| |
Collapse
|
246
|
Correa Leite PE, de Araujo Portes J, Pereira MR, Russo FB, Martins-Duarte ES, Almeida Dos Santos N, Attias M, Barrantes FJ, Baleeiro Beltrão-Braga PC, de Souza W. Morphological and biochemical repercussions of Toxoplasma gondii infection in a 3D human brain neurospheres model. Brain Behav Immun Health 2021; 11:100190. [PMID: 34589727 PMCID: PMC8474451 DOI: 10.1016/j.bbih.2020.100190] [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: 12/06/2020] [Accepted: 12/06/2020] [Indexed: 12/19/2022] Open
Abstract
Background Toxoplasmosis is caused by the parasite Toxoplasma gondii that can infect the central nervous system (CNS), promoting neuroinflammation, neuronal loss, neurotransmitter imbalance and behavioral alterations. T. gondii infection is also related to neuropsychiatric disorders such as schizophrenia. The pathogenicity and inflammatory response in rodents are different to the case of humans, compromising the correlation between the behavioral alterations and physiological modifications observed in the disease. In the present work we used BrainSpheres, a 3D CNS model derived from human pluripotent stem cells (iPSC), to investigate the morphological and biochemical repercussions of T. gondii infection in human neural cells. Methods We evaluated T. gondii ME49 strain proliferation and cyst formation in both 2D cultured human neural cells and BrainSpheres. Aspects of cell morphology, ultrastructure, viability, gene expression of neural phenotype markers, as well as secretion of inflammatory mediators were evaluated for 2 and 4 weeks post infection in BrainSpheres. Results T. gondii can infect BrainSpheres, proliferating and inducing cysts formation, neural cell death, alteration in neural gene expression and triggering the release of several inflammatory mediators. Conclusions BrainSpheres reproduce many aspects of T. gondii infection in human CNS, constituting a useful model to study the neurotoxicity and neuroinflammation mediated by the parasite. In addition, these data could be important for future studies aiming at better understanding possible correlations between psychiatric disorders and human CNS infection with T. gondii. T. gondii infects, proliferates and induce cysts formation in neurospheres. T. gondii infection induces neural cell death in neurospheres. T. gondii infection promotes alteration in neural gene expression in neurospheres. T. gondii infection promotes release of inflammatory mediators in neurospheres.
Collapse
Affiliation(s)
- Paulo Emilio Correa Leite
- Institute of Biophysics Carlos Chagas Filho and National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, RJ, Brazil.,Directory of Metrology Applied to Life Sciences (Dimav), National Institute of Metrology Quality and Technology (INMETRO), Duque de Caxias, RJ, Brazil
| | - Juliana de Araujo Portes
- Institute of Biophysics Carlos Chagas Filho and National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, RJ, Brazil
| | | | - Fabiele Baldino Russo
- Laboratory of Disease Modeling, Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| | - Erica S Martins-Duarte
- Institute of Biophysics Carlos Chagas Filho and National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, RJ, Brazil.,Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Nathalia Almeida Dos Santos
- Laboratory of Disease Modeling, Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil.,Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, UK
| | - Marcia Attias
- Institute of Biophysics Carlos Chagas Filho and National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, RJ, Brazil
| | - Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Institute for Biomedical Research (BIOMED), UCA-CONICET, Buenos Aires, Argentina
| | - Patricia Cristina Baleeiro Beltrão-Braga
- Laboratory of Disease Modeling, Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil.,Scientific Platform Pasteur-USP, São Paulo, SP, Brazil
| | - Wanderley de Souza
- Institute of Biophysics Carlos Chagas Filho and National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, RJ, Brazil
| |
Collapse
|
247
|
Basnayake K, Mazaud D, Kushnireva L, Bemelmans A, Rouach N, Korkotian E, Holcman D. Nanoscale molecular architecture controls calcium diffusion and ER replenishment in dendritic spines. SCIENCE ADVANCES 2021; 7:eabh1376. [PMID: 34524854 PMCID: PMC8443180 DOI: 10.1126/sciadv.abh1376] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/19/2021] [Indexed: 06/13/2023]
Abstract
Dendritic spines are critical components of neuronal synapses as they receive and transform synaptic inputs into a succession of calcium-regulated biochemical events. The spine apparatus (SA), an extension of smooth endoplasmic reticulum, regulates slow and fast calcium dynamics in spines. Calcium release events deplete SA calcium ion reservoir rapidly, yet the next cycle of signaling requires its replenishment. How spines achieve this replenishment without triggering calcium release remains unclear. Using computational modeling, calcium and STED superresolution imaging, we show that the SA replenishment involves the store-operated calcium entry pathway during spontaneous calcium transients. We identified two main conditions for SA replenishment without depletion: a small amplitude and a slow timescale for calcium influx, and a close proximity between SA and plasma membranes. Thereby, spine’s nanoscale organization separates SA replenishment from depletion. We further conclude that spine’s receptor organization also determines the calcium dynamics during the induction of long-term synaptic changes.
Collapse
Affiliation(s)
- Kanishka Basnayake
- Computational Biology and Applied Mathematics, Institut de Biologie de l’École Normale Supérieure-PSL, Paris, France
| | - David Mazaud
- Neuroglial Interactions in Cerebral Physiology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | | | - Alexis Bemelmans
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de biologie François Jacob, Molecular Imaging Research Center and Centre National de la Recherche Scientifique UMR9199, Université Paris-Sud, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Eduard Korkotian
- Faculty of Biology, Perm State University, Perm, Russia
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - David Holcman
- Computational Biology and Applied Mathematics, Institut de Biologie de l’École Normale Supérieure-PSL, Paris, France
- Churchill College and the Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK
| |
Collapse
|
248
|
Shiota Y, Matsudaira I, Takeuchi H, Ono C, Tomita H, Kawashima R, Taki Y. The influence of NRXN1 on systemizing and the brain structure in healthy adults. Brain Imaging Behav 2021; 16:692-701. [PMID: 34529206 DOI: 10.1007/s11682-021-00530-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2021] [Indexed: 11/24/2022]
Abstract
Certain behavioral characteristics of autism spectrum disorder can be found in otherwise healthy people. Individuals with difficulties in social adaptation may have subclinical autistic traits; however, effective biomarkers of these traits have not yet been established. There is a dire need for objective indices of these traits that combine behavior, brain images, and genetic information. In this study, we examined the association among a single nucleotide polymorphism of NRXN1 (rs858932; C/G), autistic traits, and brain structure in 311 healthy adults. We found that carriers of minor alleles (carriers of the G-allele) had significantly higher systemizing scores than major-allele (C-allele) homozygotes. Furthermore, the regional white matter volume in the right anterior limb of the internal capsule was significantly greater in carriers of the G-allele than in C-allele homozygotes. To the best of our knowledge, this is the first report of NRXN1 rs858932 being involved in systemizing and the brain structure of healthy adults. Our findings provide insight into the effects of genetics on autistic traits and their respective neural substrates.
Collapse
Affiliation(s)
- Yuka Shiota
- Department of Nuclear Medicine and Radiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Izumi Matsudaira
- Smart-Aging Research Center, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
| | - Hikaru Takeuchi
- Division of Developmental Cognitive Neuroscience, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Chiaki Ono
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Hiroaki Tomita
- Department of Disaster Psychiatry, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan.,Department of Psychiatry, Tohoku University Hospital, Sendai, Japan
| | - Ryuta Kawashima
- Division of Developmental Cognitive Neuroscience, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.,Department of Advanced Brain Science, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.,Smart-Aging Research Center, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Yasuyuki Taki
- Department of Nuclear Medicine and Radiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.,Smart-Aging Research Center, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
249
|
Gawlińska K, Gawliński D, Borczyk M, Korostyński M, Przegaliński E, Filip M. A Maternal High-Fat Diet during Early Development Provokes Molecular Changes Related to Autism Spectrum Disorder in the Rat Offspring Brain. Nutrients 2021; 13:3212. [PMID: 34579089 PMCID: PMC8467420 DOI: 10.3390/nu13093212] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 12/23/2022] Open
Abstract
Autism spectrum disorder (ASD) is a disruptive neurodevelopmental disorder manifested by abnormal social interactions, communication, emotional circuits, and repetitive behaviors and is more often diagnosed in boys than in girls. It is postulated that ASD is caused by a complex interaction between genetic and environmental factors. Epigenetics provides a mechanistic link between exposure to an unbalanced maternal diet and persistent modifications in gene expression levels that can lead to phenotype changes in the offspring. To better understand the impact of the early development environment on the risk of ASD in offspring, we assessed the effect of maternal high-fat (HFD), high-carbohydrate, and mixed diets on molecular changes in adolescent and young adult offspring frontal cortex and hippocampus. Our results showed that maternal HFD significantly altered the expression of 48 ASD-related genes in the frontal cortex of male offspring. Moreover, exposure to maternal HFD led to sex- and age-dependent changes in the protein levels of ANKRD11, EIF4E, NF1, SETD1B, SHANK1 and TAOK2, as well as differences in DNA methylation levels in the frontal cortex and hippocampus of the offspring. Taken together, it was concluded that a maternal HFD during pregnancy and lactation periods can lead to abnormal brain development within the transcription and translation of ASD-related genes mainly in male offspring.
Collapse
Affiliation(s)
- Kinga Gawlińska
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna Street 12, 31-343 Kraków, Poland; (K.G.); (E.P.); (M.F.)
| | - Dawid Gawliński
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna Street 12, 31-343 Kraków, Poland; (K.G.); (E.P.); (M.F.)
| | - Małgorzata Borczyk
- Maj Institute of Pharmacology Polish Academy of Sciences, Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Smętna Street 12, 31-343 Kraków, Poland; (M.B.); (M.K.)
| | - Michał Korostyński
- Maj Institute of Pharmacology Polish Academy of Sciences, Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Smętna Street 12, 31-343 Kraków, Poland; (M.B.); (M.K.)
| | - Edmund Przegaliński
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna Street 12, 31-343 Kraków, Poland; (K.G.); (E.P.); (M.F.)
| | - Małgorzata Filip
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna Street 12, 31-343 Kraków, Poland; (K.G.); (E.P.); (M.F.)
| |
Collapse
|
250
|
Watanabe S, Kurotani T, Oga T, Noguchi J, Isoda R, Nakagami A, Sakai K, Nakagaki K, Sumida K, Hoshino K, Saito K, Miyawaki I, Sekiguchi M, Wada K, Minamimoto T, Ichinohe N. Functional and molecular characterization of a non-human primate model of autism spectrum disorder shows similarity with the human disease. Nat Commun 2021; 12:5388. [PMID: 34526497 PMCID: PMC8443557 DOI: 10.1038/s41467-021-25487-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 08/12/2021] [Indexed: 02/08/2023] Open
Abstract
Autism spectrum disorder (ASD) is a multifactorial disorder with characteristic synaptic and gene expression changes. Early intervention during childhood is thought to benefit prognosis. Here, we examined the changes in cortical synaptogenesis, synaptic function, and gene expression from birth to the juvenile stage in a marmoset model of ASD induced by valproic acid (VPA) treatment. Early postnatally, synaptogenesis was reduced in this model, while juvenile-age VPA-treated marmosets showed increased synaptogenesis, similar to observations in human tissue. During infancy, synaptic plasticity transiently increased and was associated with altered vocalization. Synaptogenesis-related genes were downregulated early postnatally. At three months of age, the differentially expressed genes were associated with circuit remodeling, similar to the expression changes observed in humans. In summary, we provide a functional and molecular characterization of a non-human primate model of ASD, highlighting its similarity to features observed in human ASD.
Collapse
Affiliation(s)
- Satoshi Watanabe
- grid.419280.60000 0004 1763 8916Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan
| | - Tohru Kurotani
- grid.419280.60000 0004 1763 8916Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan
| | - Tomofumi Oga
- grid.419280.60000 0004 1763 8916Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan
| | - Jun Noguchi
- grid.419280.60000 0004 1763 8916Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan
| | - Risa Isoda
- grid.419280.60000 0004 1763 8916Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan
| | - Akiko Nakagami
- grid.419280.60000 0004 1763 8916Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan ,grid.411827.90000 0001 2230 656XDepartment of Psychology, Japan Women’s University, Kawasaki, Kanagawa Japan
| | - Kazuhisa Sakai
- grid.419280.60000 0004 1763 8916Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan
| | - Keiko Nakagaki
- grid.419280.60000 0004 1763 8916Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan
| | - Kayo Sumida
- grid.459996.e0000 0004 0376 2692Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Konohana-ku, Osaka, Japan
| | - Kohei Hoshino
- grid.417741.00000 0004 1797 168XPreclinical Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Konohana-ku, Osaka, Japan
| | - Koichi Saito
- grid.459996.e0000 0004 0376 2692Environmental Health Science Laboratory, Sumitomo Chemical Co., Ltd., Konohana-ku, Osaka, Japan
| | - Izuru Miyawaki
- grid.417741.00000 0004 1797 168XPreclinical Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Konohana-ku, Osaka, Japan
| | - Masayuki Sekiguchi
- grid.419280.60000 0004 1763 8916Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan
| | - Keiji Wada
- grid.419280.60000 0004 1763 8916Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan
| | - Takafumi Minamimoto
- grid.482503.80000 0004 5900 003XDepartment of Functional Brain Imaging, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba, Japan
| | - Noritaka Ichinohe
- grid.419280.60000 0004 1763 8916Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo Japan
| |
Collapse
|