1
|
Kovlyagina I, Wierczeiko A, Todorov H, Jacobi E, Tevosian M, von Engelhardt J, Gerber S, Lutz B. Leveraging interindividual variability in threat conditioning of inbred mice to model trait anxiety. PLoS Biol 2024; 22:e3002642. [PMID: 38805548 PMCID: PMC11161093 DOI: 10.1371/journal.pbio.3002642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/07/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
Trait anxiety is a major risk factor for stress-induced and anxiety disorders in humans. However, animal models accounting for the interindividual variability in stress vulnerability are largely lacking. Moreover, the pervasive bias of using mostly male animals in preclinical studies poorly reflects the increased prevalence of psychiatric disorders in women. Using the threat imminence continuum theory, we designed and validated an auditory aversive conditioning-based pipeline in both female and male mice. We operationalised trait anxiety by harnessing the naturally occurring variability of defensive freezing responses combined with a model-based clustering strategy. While sustained freezing during prolonged retrieval sessions was identified as an anxiety-endophenotype behavioral marker in both sexes, females were consistently associated with an increased freezing response. RNA-sequencing of CeA, BLA, ACC, and BNST revealed massive differences in phasic and sustained responders' transcriptomes, correlating with transcriptomic signatures of psychiatric disorders, particularly post-traumatic stress disorder (PTSD). Moreover, we detected significant alterations in the excitation/inhibition balance of principal neurons in the lateral amygdala. These findings provide compelling evidence that trait anxiety in inbred mice can be leveraged to develop translationally relevant preclinical models to investigate mechanisms of stress susceptibility in a sex-specific manner.
Collapse
Affiliation(s)
- Irina Kovlyagina
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Anna Wierczeiko
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Hristo Todorov
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Eric Jacobi
- Institute of Pathophysiology, and Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Margarita Tevosian
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
| | - Jakob von Engelhardt
- Institute of Pathophysiology, and Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
| |
Collapse
|
2
|
Molecular and cellular evolution of the amygdala across species analyzed by single-nucleus transcriptome profiling. Cell Discov 2023; 9:19. [PMID: 36788214 PMCID: PMC9929086 DOI: 10.1038/s41421-022-00506-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/24/2022] [Indexed: 02/16/2023] Open
Abstract
The amygdala, or an amygdala-like structure, is found in the brains of all vertebrates and plays a critical role in survival and reproduction. However, the cellular architecture of the amygdala and how it has evolved remain elusive. Here, we generated single-nucleus RNA-sequencing data for more than 200,000 cells in the amygdala of humans, macaques, mice, and chickens. Abundant neuronal cell types from different amygdala subnuclei were identified in all datasets. Cross-species analysis revealed that inhibitory neurons and inhibitory neuron-enriched subnuclei of the amygdala were well-conserved in cellular composition and marker gene expression, whereas excitatory neuron-enriched subnuclei were relatively divergent. Furthermore, LAMP5+ interneurons were much more abundant in primates, while DRD2+ inhibitory neurons and LAMP5+SATB2+ excitatory neurons were dominant in the human central amygdalar nucleus (CEA) and basolateral amygdalar complex (BLA), respectively. We also identified CEA-like neurons and their species-specific distribution patterns in chickens. This study highlights the extreme cell-type diversity in the amygdala and reveals the conservation and divergence of cell types and gene expression patterns across species that may contribute to species-specific adaptations.
Collapse
|
3
|
Pawlak P, Burren A, Seitz A, Pietsch C. Effects of different acute stressors on the regulation of appetite genes in the carp ( Cyprinus carpio L.) brain. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230040. [PMID: 36816841 PMCID: PMC9929511 DOI: 10.1098/rsos.230040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 06/18/2023]
Abstract
Our understanding of the timing of stress responses and specific roles of different regulatory pathways that drive stress responses is incomplete. In particular, the regulation of appetite genes as a consequence of exposure to different stressors has not been studied in sufficient detail in fish. Therefore, a stress trial was conducted with koi carp, aiming at identifying typical effects of stress on regulation of appetite genes. The stressors tank manipulation, air exposure and feed rewarding were chosen. The responses to these stressors were evaluated 10, 30 and 60 min after the stressors were applied. Orexigenic and anorexigenic genes were investigated in four different brain regions (telencephalon, hypothalamus, optic tectum and rhombencephalon). The results show that, apart from the typical appetite regulation in the hypothalamus, the different brain regions also display pronounced responses of appetite genes to the different stressors. In addition, several genes in the serotonergic, dopaminergic and gaba-related pathways were investigated. These genes revealed that rearing in pairs of two and opening of the tank lid affected anorexigenic genes, such as cart and cck, which were not changed by air exposure or feed rewarding. Moreover, distress and eustress led to limited, but distinguishable gene expression pattern changes in the investigated brain regions.
Collapse
Affiliation(s)
- Paulina Pawlak
- Agronomy, Bern University of Applied Sciences, Zollikofen, Bern CH-2052, Switzerland
- Division of Behavioural Ecology, Institute of Ecology and Evolution, University of Bern, Wohlenstrasse 50a, CH-3032, Hinterkappelen, Bern, Switzerland
| | - Alexander Burren
- Agronomy, Bern University of Applied Sciences, Zollikofen, Bern CH-2052, Switzerland
| | - Andreas Seitz
- Institute of Natural Resource Sciences, Zurich University of Applied Sciences, Wädenswil, Zürich CH-8820, Switzerland
| | - Constanze Pietsch
- Agronomy, Bern University of Applied Sciences, Zollikofen, Bern CH-2052, Switzerland
| |
Collapse
|
4
|
Kim YK, Jung YS, Song J. Transcriptome Profile in the Mouse Brain of Hepatic Encephalopathy and Alzheimer's Disease. Int J Mol Sci 2022; 24:ijms24010675. [PMID: 36614117 PMCID: PMC9821016 DOI: 10.3390/ijms24010675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/13/2022] [Accepted: 12/18/2022] [Indexed: 01/03/2023] Open
Abstract
Hepatic encephalopathy (HE) is a chronic metabolic disease accompanied by neuropathological and neuropsychiatric features, including memory deficits, psychomotor dysfunction, depression, and anxiety. Alzheimer's disease (AD), the most common neurodegenerative disease, is characterized by tau hyperphosphorylation, excessive amyloid beta (Aβ) accumulation, the formation of fibrillary tangles, hippocampus atrophy, and neuroinflammation. Recent studies have suggested a positive correlation between HE and AD. Some studies reported that an impaired cholesterol pathway, abnormal bile acid secretion, excessive ammonia level, impaired Aβ clearance, astrocytic dysfunction, and abnormal γ-aminobutyric acid GABAergic neuronal signaling in HE may also be involved in AD pathology. However, the mechanisms and related genes involved in AD-like pathology in the HE brain are unclear. Thus, we compared the cortical transcriptome profile between an HE mouse model, bile duct ligation (BDL), and an AD mouse model, the 5×FAD. Our study showed that the expression of many genes implicated in HE is associated with neuronal dysfunction in AD mice. We found changes in various protein-coding RNAs, implicated in synapses, neurogenesis, neuron projection, neuron differentiation, and neurite outgrowth, and non-coding RNAs possibly associated with neuropathology. Our data provide an important resource for further studies to elucidate AD-like pathophysiology in HE patients.
Collapse
Affiliation(s)
- Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea
| | - Yoon Seok Jung
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea
- Correspondence: ; Tel.: +82-61-379-2706; Fax: +82-61-375-5834
| |
Collapse
|
5
|
Li Z, Sun Y, Ding L, Yang J, Huang J, Cheng M, Wu L, Zhuang Z, Chen C, Huang Y, Zhu Z, Jiang S, Huang F, Wang C, Liu S, Liu L, Lei Y. Deciphering the distinct transcriptomic and gene regulatory map in adult macaque basal ganglia cells. Gigascience 2022; 12:giad095. [PMID: 38091510 PMCID: PMC10716911 DOI: 10.1093/gigascience/giad095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 08/09/2023] [Accepted: 10/10/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The basal ganglia are a complex of interconnected subcortical structures located beneath the mammalian cerebral cortex. The degeneration of dopaminergic neurons in the basal ganglia is the primary pathological feature of Parkinson's disease. Due to a lack of integrated analysis of multiomics datasets across multiple basal ganglia brain regions, very little is known about the regulatory mechanisms of this area. FINDINGS We utilized high-throughput transcriptomic and epigenomic analysis to profile over 270,000 single-nucleus cells to create a cellular atlas of the basal ganglia, characterizing the cellular composition of 4 regions of basal ganglia in adult macaque brain, including the striatum, substantia nigra (SN), globus pallidum, and amygdala. We found a distinct epigenetic regulation on gene expression of neuronal and nonneuronal cells across regions in basal ganglia. We identified a cluster of SN-specific astrocytes associated with neurodegenerative diseases and further explored the conserved and primate-specific transcriptomics in SN cell types across human, macaque, and mouse. Finally, we integrated our epigenetic landscape of basal ganglia cells with human disease heritability and identified a regulatory module consisting of candidate cis-regulatory elements that are specific to medium spiny neurons and associated with schizophrenia. CONCLUSIONS In general, our macaque basal ganglia atlas provides valuable insights into the comprehensive transcriptome and epigenome of the most important and populous cell populations in the macaque basal ganglia. We have identified 49 cell types based on transcriptomic profiles and 47 cell types based on epigenomic profiles, some of which exhibit region specificity, and characterized the molecular relationships underlying these brain regions.
Collapse
Affiliation(s)
- Zihao Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | - Yunong Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | | | - Jing Yang
- BGI Research, Hangzhou 310030, China
| | | | | | - Liang Wu
- BGI Research, Shenzhen 518083, China
| | | | - Cheng Chen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | - Yunqi Huang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | - Zhiyong Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | - Siyuan Jiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | - Fubaoqian Huang
- BGI Research, Hangzhou 310030, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Chunqing Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Shenzhen 518083, China
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
| | - Longqi Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
| | - Ying Lei
- BGI Research, Shenzhen 518083, China
| |
Collapse
|
6
|
Sakpichaisakul K, Boonkrongsak R, Lertbutsayanukul P, Iemwimangsa N, Klumsathian S, Panthan B, Trachoo O. Epileptic spasms related to neuronal differentiation factor 2 (NEUROD2) mutation respond to combined vigabatrin and high dose prednisolone therapy. BMC Neurol 2022; 22:461. [PMID: 36494631 PMCID: PMC9733267 DOI: 10.1186/s12883-022-02992-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Epileptic spasms are a devastating form of early infantile epileptic encephalopathy (EIEE) with various etiologies. Early diagnosis and a shorter lead time to treatment are crucial to stop the seizures and optimize the neurodevelopmental outcome. Genetic testing has become an integral part of epilepsy care that directly guides management and family planning and discovers new targeted treatments. Neuronal differentiation Factor 2 (NEUROD2) variants have recently been a cause of neurodevelopmental disorders (NDDs) and EIEEs with distinctive features. However, there is limited information about the clinical and electroencephalographic response of epileptic spasm treatment in NEUROD2-related NDD syndrome. CASE PRESENTATION We report a female patient of Southeast Asian ethnicity with global developmental delay and epileptic spasms commencing in the first few months of life. A novel de novo heterozygous pathogenic NEUROD2 variant, p. E130Q, was subsequently identified by whole-exome sequencing. Electroencephalogram before treatment showed multifocal independent spikes predominantly in both posterior head regions and demonstrated marked improvement following combined vigabatrin and high-dose prednisolone treatment. However, multiple courses of relapse occurred after weaning off the antiseizure medication. CONCLUSIONS We propose that epileptic spasms related to de novo NEUROD2 pathogenic variant respond well to combined vigabatrin and high-dose prednisolone therapy. These findings may imply the benefit of using combination therapy to treat epileptic spasms in NEUROD2-related NDD syndrome.
Collapse
Affiliation(s)
- Kullasate Sakpichaisakul
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, College of Medicine, Rangsit University, Bangkok, 10400, Thailand
| | - Rachata Boonkrongsak
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, College of Medicine, Rangsit University, Bangkok, 10400, Thailand
| | | | - Nareenart Iemwimangsa
- Centre for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Sommon Klumsathian
- Centre for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | | | - Objoon Trachoo
- Centre for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand.
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama 6 Road, Ratchathewi, Bangkok, 10400, Thailand.
| |
Collapse
|
7
|
Leung HW, Foo G, VanDongen A. Arc Regulates Transcription of Genes for Plasticity, Excitability and Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081946. [PMID: 36009494 PMCID: PMC9405677 DOI: 10.3390/biomedicines10081946] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
The immediate early gene Arc is a master regulator of synaptic function and a critical determinant of memory consolidation. Here, we show that Arc interacts with dynamic chromatin and closely associates with histone markers for active enhancers and transcription in cultured rat hippocampal neurons. Both these histone modifications, H3K27Ac and H3K9Ac, have recently been shown to be upregulated in late-onset Alzheimer’s disease (AD). When Arc induction by pharmacological network activation was prevented using a short hairpin RNA, the expression profile was altered for over 1900 genes, which included genes associated with synaptic function, neuronal plasticity, intrinsic excitability, and signalling pathways. Interestingly, about 100 Arc-dependent genes are associated with the pathophysiology of AD. When endogenous Arc expression was induced in HEK293T cells, the transcription of many neuronal genes was increased, suggesting that Arc can control expression in the absence of activated signalling pathways. Taken together, these data establish Arc as a master regulator of neuronal activity-dependent gene expression and suggest that it plays a significant role in the pathophysiology of AD.
Collapse
Affiliation(s)
| | - Gabriel Foo
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Antonius VanDongen
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Correspondence:
| |
Collapse
|
8
|
The HDAC inhibitor CI-994 acts as a molecular memory aid by facilitating synaptic and intracellular communication after learning. Proc Natl Acad Sci U S A 2022; 119:e2116797119. [PMID: 35613054 PMCID: PMC9295763 DOI: 10.1073/pnas.2116797119] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Memory formation relies on a plethora of functions, including epigenetic modifications. Over recent years, multiple studies have indicated the potential of HDAC inhibitors (HDACis) as cognitive enhancers, but their mode of action is not fully understood. Here, we tested whether HDACi treatment improves memory formation via “cognitive epigenetic priming,” stipulating that HDACis—without inherent target specificity—specifically enhance naturally occurring plasticity processes. We found that combining HDACis with fear learning, but not either treatment alone, enhances synaptic plasticity as well as memory-promoting transcriptional signaling in the hippocampus, a brain area recruited by fear learning, but not in unrelated areas. These results lend experimental support to the theory of cognitive epigenetic priming. Long-term memory formation relies on synaptic plasticity, neuronal activity-dependent gene transcription, and epigenetic modifications. Multiple studies have shown that HDAC inhibitor (HDACi) treatments can enhance individual aspects of these processes and thereby act as putative cognitive enhancers. However, their mode of action is not fully understood. In particular, it is unclear how systemic application of HDACis, which are devoid of substrate specificity, can target pathways that promote memory formation. In this study, we explore the electrophysiological, transcriptional, and epigenetic responses that are induced by CI-994, a class I HDACi, combined with contextual fear conditioning (CFC) in mice. We show that CI-994–mediated improvement of memory formation is accompanied by enhanced long-term potentiation in the hippocampus, a brain region recruited by CFC, but not in the striatum, a brain region not primarily implicated in fear learning. Furthermore, using a combination of bulk and single-cell RNA-sequencing, we find that, when paired with CFC, HDACi treatment engages synaptic plasticity-promoting gene expression more strongly in the hippocampus, specifically in the dentate gyrus (DG). Finally, using chromatin immunoprecipitation-sequencing (ChIP-seq) of DG neurons, we show that the combined action of HDACi application and conditioning is required to elicit enhancer histone acetylation in pathways that underlie improved memory performance. Together, these results indicate that systemic HDACi administration amplifies brain region-specific processes that are naturally induced by learning.
Collapse
|
9
|
Tutukova S, Tarabykin V, Hernandez-Miranda LR. The Role of Neurod Genes in Brain Development, Function, and Disease. Front Mol Neurosci 2021; 14:662774. [PMID: 34177462 PMCID: PMC8221396 DOI: 10.3389/fnmol.2021.662774] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/11/2021] [Indexed: 01/14/2023] Open
Abstract
Transcriptional regulation is essential for the correct functioning of cells during development and in postnatal life. The basic Helix-loop-Helix (bHLH) superfamily of transcription factors is well conserved throughout evolution and plays critical roles in tissue development and tissue maintenance. A subgroup of this family, called neural lineage bHLH factors, is critical in the development and function of the central nervous system. In this review, we will focus on the function of one subgroup of neural lineage bHLH factors, the Neurod family. The Neurod family has four members: Neurod1, Neurod2, Neurod4, and Neurod6. Available evidence shows that these four factors are key during the development of the cerebral cortex but also in other regions of the central nervous system, such as the cerebellum, the brainstem, and the spinal cord. We will also discuss recent reports that link the dysfunction of these transcription factors to neurological disorders in humans.
Collapse
Affiliation(s)
- Svetlana Tutukova
- Institute of Neuroscience, Lobachevsky University of Nizhny Novgorod, Nizhny Novgorod, Russia.,Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute for Cell- and Neurobiology, Berlin, Germany
| | - Victor Tarabykin
- Institute of Neuroscience, Lobachevsky University of Nizhny Novgorod, Nizhny Novgorod, Russia.,Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute for Cell- and Neurobiology, Berlin, Germany
| | - Luis R Hernandez-Miranda
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute for Cell- and Neurobiology, Berlin, Germany
| |
Collapse
|
10
|
Safari MS, Obexer D, Baier-Bitterlich G, Zur Nedden S. PKN1 Is a Novel Regulator of Hippocampal GluA1 Levels. Front Synaptic Neurosci 2021; 13:640495. [PMID: 33613259 PMCID: PMC7892898 DOI: 10.3389/fnsyn.2021.640495] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/18/2021] [Indexed: 11/13/2022] Open
Abstract
Alterations in the processes that control α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) expression, assembly and trafficking are closely linked to psychiatric and neurodegenerative disorders. We have recently shown that the serine/threonine kinase Protein kinase N1 (PKN1) is a developmentally active regulator of cerebellar synaptic maturation by inhibiting AKT and the neurogenic transcription factor neurogenic differentiation factor-2 (NeuroD2). NeuroD2 is involved in glutamatergic synaptic maturation by regulating expression levels of various synaptic proteins. Here we aimed to study the effect of Pkn1 knockout on AKT phosphorylation and NeuroD2 levels in the hippocampus and the subsequent expression levels of the NeuroD2 targets and AMPAR subunits: glutamate receptor 1 (GluA1) and GluA2/3. We show that PKN1 is expressed throughout the hippocampus. Interestingly, not only postnatal but also adult hippocampal phospho-AKT and NeuroD2 levels were significantly elevated upon Pkn1 knockout. Postnatal and adult Pkn1 -/- hippocampi showed enhanced expression of the AMPAR subunit GluA1, particularly in area CA1. Surprisingly, GluA2/3 levels were not different between both genotypes. In addition to higher protein levels, we also found an enhanced GluA1 content in the membrane fraction of postnatal and adult Pkn1 -/- animals, while GluA2/3 levels remained unchanged. This points toward a very specific regulation of GluA1 expression and/or trafficking by the novel PKN1-AKT-NeuroD2 axis. Considering the important role of GluA1 in hippocampal development as well as the pathophysiology of several disorders, ranging from Alzheimer's, to depression and schizophrenia, our results validate PKN1 for future studies into neurological disorders related to altered AMPAR subunit expression in the hippocampus.
Collapse
Affiliation(s)
- Motahareh Solina Safari
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, Innsbruck, Austria
| | - Dido Obexer
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Stephanie Zur Nedden
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Disruption of NEUROD2 causes a neurodevelopmental syndrome with autistic features via cell-autonomous defects in forebrain glutamatergic neurons. Mol Psychiatry 2021; 26:6125-6148. [PMID: 34188164 PMCID: PMC8760061 DOI: 10.1038/s41380-021-01179-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 05/17/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023]
Abstract
While the transcription factor NEUROD2 has recently been associated with epilepsy, its precise role during nervous system development remains unclear. Using a multi-scale approach, we set out to understand how Neurod2 deletion affects the development of the cerebral cortex in mice. In Neurod2 KO embryos, cortical projection neurons over-migrated, thereby altering the final size and position of layers. In juvenile and adults, spine density and turnover were dysregulated in apical but not basal compartments in layer 5 neurons. Patch-clamp recordings in layer 5 neurons of juvenile mice revealed increased intrinsic excitability. Bulk RNA sequencing showed dysregulated expression of many genes associated with neuronal excitability and synaptic function, whose human orthologs were strongly associated with autism spectrum disorders (ASD). At the behavior level, Neurod2 KO mice displayed social interaction deficits, stereotypies, hyperactivity, and occasionally spontaneous seizures. Mice heterozygous for Neurod2 had similar defects, indicating that Neurod2 is haploinsufficient. Finally, specific deletion of Neurod2 in forebrain excitatory neurons recapitulated cellular and behavioral phenotypes found in constitutive KO mice, revealing the region-specific contribution of dysfunctional Neurod2 in symptoms. Informed by these neurobehavioral features in mouse mutants, we identified eleven patients from eight families with a neurodevelopmental disorder including intellectual disability and ASD associated with NEUROD2 pathogenic mutations. Our findings demonstrate crucial roles for Neurod2 in neocortical development, whose alterations can cause neurodevelopmental disorders including intellectual disability and ASD.
Collapse
|
12
|
Runge K, Cardoso C, de Chevigny A. Dendritic Spine Plasticity: Function and Mechanisms. Front Synaptic Neurosci 2020. [DOI: 10.3389/fnsyn.2020.00036
expr 823669561 + 872784217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
|
13
|
Runge K, Cardoso C, de Chevigny A. Dendritic Spine Plasticity: Function and Mechanisms. Front Synaptic Neurosci 2020; 12:36. [PMID: 32982715 PMCID: PMC7484486 DOI: 10.3389/fnsyn.2020.00036] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Dendritic spines are small protrusions studding neuronal dendrites, first described in 1888 by Ramón y Cajal using his famous Golgi stainings. Around 50 years later the advance of electron microscopy (EM) confirmed Cajal's intuition that spines constitute the postsynaptic site of most excitatory synapses in the mammalian brain. The finding that spine density decreases between young and adult ages in fixed tissues suggested that spines are dynamic. It is only a decade ago that two-photon microscopy (TPM) has unambiguously proven the dynamic nature of spines, through the repeated imaging of single spines in live animals. Spine dynamics comprise formation, disappearance, and stabilization of spines and are modulated by neuronal activity and developmental age. Here, we review several emerging concepts in the field that start to answer the following key questions: What are the external signals triggering spine dynamics and the molecular mechanisms involved? What is, in return, the role of spine dynamics in circuit-rewiring, learning, and neuropsychiatric disorders?
Collapse
Affiliation(s)
- Karen Runge
- Institut de Neurobiologie de la Méditerranée (INMED) INSERM U1249, Aix-Marseille University, Marseille, France
| | - Carlos Cardoso
- Institut de Neurobiologie de la Méditerranée (INMED) INSERM U1249, Aix-Marseille University, Marseille, France
| | - Antoine de Chevigny
- Institut de Neurobiologie de la Méditerranée (INMED) INSERM U1249, Aix-Marseille University, Marseille, France
| |
Collapse
|
14
|
Bisphenol a Exposure in Utero Disrupts Hypothalamic Gene Expression Particularly Genes Suspected in Autism Spectrum Disorders and Neuron and Hormone Signaling. Int J Mol Sci 2020; 21:ijms21093129. [PMID: 32365465 PMCID: PMC7246794 DOI: 10.3390/ijms21093129] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/23/2020] [Accepted: 04/27/2020] [Indexed: 01/09/2023] Open
Abstract
Bisphenol A (BPA) is an endocrine-disrupting compound detected in the urine of more than 92% of humans, easily crosses the placental barrier, and has been shown to influence gene expression during fetal brain development. The purpose of this study was to investigate the effect of in utero BPA exposure on gene expression in the anterior hypothalamus, the basal nucleus of the stria terminalis (BNST), and hippocampus in C57BL/6 mice. Mice were exposed in utero to human-relevant doses of BPA, and then RNA sequencing was performed on male PND 28 tissue from whole hypothalamus (n = 3/group) that included the medial preoptic area (mPOA) and BNST to determine whether any genes were differentially expressed between BPA-exposed and control mice. A subset of genes was selected for further study using RT-qPCR on adult tissue from hippocampus to determine whether any differentially expressed genes (DEGs) persisted into adulthood. Two different RNA-Seq workflows indicated a total of 259 genes that were differentially expressed between BPA-exposed and control mice. Gene ontology analysis indicated that those DEGs were overrepresented in categories relating to mating, cell-cell signaling, behavior, neurodevelopment, neurogenesis, synapse formation, cognition, learning behaviors, hormone activity, and signaling receptor activity, among others. Ingenuity Pathway Analysis was used to interrogate novel gene networks and upstream regulators, indicating the top five upstream regulators as huntingtin, beta-estradiol, alpha-synuclein, Creb1, and estrogen receptor (ER)-alpha. In addition, 15 DE genes were identified that are suspected in autism spectrum disorders.
Collapse
|
15
|
Koizumi R, Kiyokawa Y, Mikami K, Ishii A, Tanaka KD, Tanikawa T, Takeuchi Y. Structural differences in the brain between wild and laboratory rats (Rattus norvegicus): Potential contribution to wariness. J Vet Med Sci 2018; 80:1054-1060. [PMID: 29760315 PMCID: PMC6068309 DOI: 10.1292/jvms.18-0052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Wild animals typically exhibit defensive behaviors in response to a wider range and/or a weaker intensity of stimuli compared with domestic animals. However, little is known about the neural mechanisms underlying "wariness" in wild animals. Wild rats are one of the most accessible wild animals for experimental research. Laboratory rats are a domesticated form of wild rat, belonging to the same species, and are therefore considered suitable control animals for wild rats. Based on these factors, we analyzed structural differences in the brain between wild and laboratory rats to elucidate the neural mechanisms underlying wariness. We examined wild rats trapped in Tokyo, and weight-matched laboratory rats. We then prepared brain sections and compared the basolateral complex of the amygdala (BLA), the bed nucleus of the stria terminalis (BNST), the main olfactory bulb and the accessory olfactory bulb. The results revealed that wild rats exhibited larger BLA, BNST and caudal part of the accessory olfactory bulb compared with laboratory rats. These results suggest that the BLA, BNST, and vomeronasal system potentially contribute to wariness in wild rats.
Collapse
Affiliation(s)
- Ryoko Koizumi
- Laboratory of Veterinary Ethology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yasushi Kiyokawa
- Laboratory of Veterinary Ethology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kaori Mikami
- Laboratory of Veterinary Ethology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Akiko Ishii
- Laboratory of Veterinary Ethology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kazuyuki D Tanaka
- Technical Research Laboratory, Ikari Shodoku Corporation, 1-12-3 Akanehama, Narashino-shi, Chiba 275-0024, Japan
| | - Tsutomu Tanikawa
- Technical Research Laboratory, Ikari Shodoku Corporation, 1-12-3 Akanehama, Narashino-shi, Chiba 275-0024, Japan
| | - Yukari Takeuchi
- Laboratory of Veterinary Ethology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
16
|
Agrawal R, Garg A, Benny Malgulwar P, Sharma V, Sarkar C, Kulshreshtha R. p53 and miR-210 regulated NeuroD2, a neuronal basic helix-loop-helix transcription factor, is downregulated in glioblastoma patients and functions as a tumor suppressor under hypoxic microenvironment. Int J Cancer 2017; 142:1817-1828. [DOI: 10.1002/ijc.31209] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 10/27/2017] [Accepted: 11/29/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Rahul Agrawal
- Department of Biochemical Engineering and Biotechnology; Indian Institute of Technology Delhi; New Delhi India
| | - Ankita Garg
- Department of Biochemical Engineering and Biotechnology; Indian Institute of Technology Delhi; New Delhi India
| | - Prit Benny Malgulwar
- Department of Pathology; All India Institute of Medical Sciences; New Delhi India
| | - Vikas Sharma
- Department of Biochemical Engineering and Biotechnology; Indian Institute of Technology Delhi; New Delhi India
| | - Chitra Sarkar
- Department of Pathology; All India Institute of Medical Sciences; New Delhi India
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology; Indian Institute of Technology Delhi; New Delhi India
| |
Collapse
|
17
|
Spellmann I, Riedel M, Städtler J, Zill P, Obermeier M, Cerovecki A, Dehning S, Schennach R, Epple M, Opgen-Rhein M, Müller N, Bondy B, Möller HJ, Musil R. Associations of NEUROD2 polymorphisms and change of cognitive dysfunctions in schizophrenia and schizoaffective disorder after eight weeks of antipsychotic treatment. Cogn Neuropsychiatry 2017; 22:280-297. [PMID: 28470106 DOI: 10.1080/13546805.2017.1322502] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION NEUROD2 is a neurospecific helix-loop-helix transcription factor which has an impact on the regulation of glutamatergic and GABAergic genes. We investigated an association of NEUROD2 with neurocognitive dysfunctions in schizophrenia and schizoaffective disorder patients before and during treatment with different second-generation antipsychotics. METHODS Patients were genotyped for four different polymorphisms of the NEUROD2 gene ((rs9889354(A/G), rs1877032(C/T), rs12453682(C/T) and rs11078918(C/G)). Cognitive function was assessed at baseline and week 8. Results of individual neuropsychological tests were assigned to six cognitive domains (reaction time and quality; executive function; working, verbal and visual memory) and a general cognitive index. RESULTS 167 patients were included in the study. The NEUROD2 exonic polymorphism rs11078918 showed significant associations with verbal memory and executive functions, whereas the NEUROD2 polymorphism rs12453682 was significantly associated with working and verbal memory, executive functions and with a cognitive index. Significant associations were found at baseline and after eight weeks. Moreover, significant associations between the change in neuropsychological test results during antipsychotic treatment and the NEUROD2 polymorphisms rs11078918 and rs12453682 were observed. CONCLUSIONS Our findings suggest that the NEUROD2 gene could play a role in the pathophysiology of neurocognitive dysfunctions as well as in the change of cognitive symptoms under antipsychotic treatment in schizophrenia and schizoaffective disorder.
Collapse
Affiliation(s)
- Ilja Spellmann
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Michael Riedel
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Julia Städtler
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Peter Zill
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Michael Obermeier
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Anja Cerovecki
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Sandra Dehning
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Rebecca Schennach
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Maria Epple
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Markus Opgen-Rhein
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Norbert Müller
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Brigitta Bondy
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Hans-Jürgen Möller
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Richard Musil
- a Department of Psychiatry and Psychotherapy , Ludwig-Maximilians-University Munich , Munich , Germany
| |
Collapse
|
18
|
Chen F, Hall BJ. Synaptic activity suppresses expression of neurogenic differentiation factor 2 in an NMDA receptor-dependent manner. Synapse 2017; 71. [PMID: 28524267 DOI: 10.1002/syn.21986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/03/2017] [Accepted: 05/14/2017] [Indexed: 01/16/2023]
Abstract
Neurogenic differentiation factor 2 (NeuroD2) is a highly expressed transcription factor in the developing central nervous system. In newborn neurons, NeuroD2-mediated gene expression promotes differentiation, maturation, and survival. In addition to these early, cell-intrinsic developmental processes, NeuroD2 in postmitotic neurons also regulates synapse growth and ion channel expression to control excitability. While NeuroD2 transactivation can be induced in an activity-dependent manner, little is known about how expression of NeuroD2 itself is regulated. Using genome-wide, mRNA-based microarray analysis, we found that NeuroD2 is actually one of hundreds of genes whose mRNA levels are suppressed by synaptic activity, in a manner dependent upon N-methyl d-aspartate receptor (NMDAR) activation. We confirmed this observation both in vitro and in vivo and provide evidence that this happens at the level of transcription and not mRNA stability. Our experiments further indicate that suppression of NeuroD2 message by NMDARs likely involves both CaMKII and MAPK but not voltage-gated calcium channels, in contrast to its mechanism of transactivation. We predict from these data that NMDARs may transduce information about the level of synaptic activity a developing neuron receives, to down-regulate NeuroD2 and allow proper maturation of cortical circuits by suppressing expression of neurite and synaptic growth promoting gene products.
Collapse
Affiliation(s)
- Fading Chen
- The Department of Cell and Molecular Biology, Tulane University, School of Science and Engineering, New Orleans, Louisiana 70118
| | - Benjamin J Hall
- The Department of Cell and Molecular Biology, Tulane University, School of Science and Engineering, New Orleans, Louisiana 70118.,The Neuroscience Program, Tulane University, School of Science and Engineering, New Orleans, Louisiana 70118.,Roche Pharmaceutical Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| |
Collapse
|
19
|
Saul MC, Seward CH, Troy JM, Zhang H, Sloofman LG, Lu X, Weisner PA, Caetano-Anolles D, Sun H, Zhao SD, Chandrasekaran S, Sinha S, Stubbs L. Transcriptional regulatory dynamics drive coordinated metabolic and neural response to social challenge in mice. Genome Res 2017; 27:959-972. [PMID: 28356321 PMCID: PMC5453329 DOI: 10.1101/gr.214221.116] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 03/24/2017] [Indexed: 12/22/2022]
Abstract
Agonistic encounters are powerful effectors of future behavior, and the ability to learn from this type of social challenge is an essential adaptive trait. We recently identified a conserved transcriptional program defining the response to social challenge across animal species, highly enriched in transcription factor (TF), energy metabolism, and developmental signaling genes. To understand the trajectory of this program and to uncover the most important regulatory influences controlling this response, we integrated gene expression data with the chromatin landscape in the hypothalamus, frontal cortex, and amygdala of socially challenged mice over time. The expression data revealed a complex spatiotemporal patterning of events starting with neural signaling molecules in the frontal cortex and ending in the modulation of developmental factors in the amygdala and hypothalamus, underpinned by a systems-wide shift in expression of energy metabolism-related genes. The transcriptional signals were correlated with significant shifts in chromatin accessibility and a network of challenge-associated TFs. Among these, the conserved metabolic and developmental regulator ESRRA was highlighted for an especially early and important regulatory role. Cell-type deconvolution analysis attributed the differential metabolic and developmental signals in this social context primarily to oligodendrocytes and neurons, respectively, and we show that ESRRA is expressed in both cell types. Localizing ESRRA binding sites in cortical chromatin, we show that this nuclear receptor binds both differentially expressed energy-related and neurodevelopmental TF genes. These data link metabolic and neurodevelopmental signaling to social challenge, and identify key regulatory drivers of this process with unprecedented tissue and temporal resolution.
Collapse
Affiliation(s)
- Michael C Saul
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Christopher H Seward
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Joseph M Troy
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Illinois Informatics Institute, Urbana, Illinois 61801, USA
| | - Huimin Zhang
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Laura G Sloofman
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Xiaochen Lu
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Patricia A Weisner
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Derek Caetano-Anolles
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Hao Sun
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Sihai Dave Zhao
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Sriram Chandrasekaran
- Harvard Society of Fellows, Harvard University, Cambridge, Massachusetts 02138, USA
- Faculty of Arts and Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Saurabh Sinha
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Computer Science
- Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Lisa Stubbs
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
20
|
van Weert LTCM, Buurstede JC, Mahfouz A, Braakhuis PSM, Polman JAE, Sips HCM, Roozendaal B, Balog J, de Kloet ER, Datson NA, Meijer OC. NeuroD Factors Discriminate Mineralocorticoid From Glucocorticoid Receptor DNA Binding in the Male Rat Brain. Endocrinology 2017; 158:1511-1522. [PMID: 28324065 DOI: 10.1210/en.2016-1422] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 01/18/2017] [Indexed: 01/08/2023]
Abstract
In the limbic brain, mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs) both function as receptors for the naturally occurring glucocorticoids (corticosterone/cortisol) but mediate distinct effects on cellular physiology via transcriptional mechanisms. The transcriptional basis for specificity of these MR- vs GR-mediated effects is unknown. To address this conundrum, we have identified the extent of MR/GR DNA-binding selectivity in the rat hippocampus using chromatin immunoprecipitation followed by sequencing. We found 918 and 1450 nonoverlapping binding sites for MR and GR, respectively. Furthermore, 475 loci were co-occupied by MR and GR. De novo motif analysis resulted in a similar binding motif for both receptors at 100% of the target loci, which matched the known glucocorticoid response element (GRE). In addition, the Atoh/NeuroD consensus sequence was found in co-occurrence with all MR-specific binding sites but was absent for GR-specific or MR-GR overlapping sites. Basic helix-loop-helix family members Neurod1, Neurod2, and Neurod6 showed hippocampal expression and were hypothesized to bind the Atoh motif. Neurod2 was detected at rat hippocampal MR binding sites but not at GR-exclusive sites. All three NeuroD transcription factors acted as DNA-binding-dependent coactivators for both MR and GR in reporter assays in heterologous HEK293 cells, likely via indirect interactions with the receptors. In conclusion, a NeuroD family member binding to an additional motif near the GRE seems to drive specificity for MR over GR binding at hippocampal binding sites.
Collapse
Affiliation(s)
- Lisa T C M van Weert
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University, 6525 EN, Nijmegen, The Netherlands
| | - Jacobus C Buurstede
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Ahmed Mahfouz
- Department of Radiology, Division of Image Processing, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
- Delft Bioinformatics Laboratory, Delft University of Technology, 2628 CD, Delft, The Netherlands
| | - Pamela S M Braakhuis
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - J Annelies E Polman
- Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research, 2300 RC, Leiden, The Netherlands
| | - Hetty C M Sips
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University, 6525 EN, Nijmegen, The Netherlands
| | - Judit Balog
- Department of Human Genetics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - E Ronald de Kloet
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
- Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research, 2300 RC, Leiden, The Netherlands
| | - Nicole A Datson
- Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research, 2300 RC, Leiden, The Netherlands
| | - Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
21
|
NEUROD2 Regulates Stim1 Expression and Store-Operated Calcium Entry in Cortical Neurons. eNeuro 2017; 4:eN-NWR-0255-16. [PMID: 28303257 PMCID: PMC5343279 DOI: 10.1523/eneuro.0255-16.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 01/18/2017] [Accepted: 01/25/2017] [Indexed: 12/26/2022] Open
Abstract
Calcium signaling controls many key processes in neurons, including gene expression, axon guidance, and synaptic plasticity. In contrast to calcium influx through voltage- or neurotransmitter-gated channels, regulatory pathways that control store-operated calcium entry (SOCE) in neurons are poorly understood. Here, we report a transcriptional control of Stim1 (stromal interaction molecule 1) gene, which is a major sensor of endoplasmic reticulum (ER) calcium levels and a regulator of SOCE. By using a genome-wide chromatin immunoprecipitation and sequencing approach in mice, we find that NEUROD2, a neurogenic transcription factor, binds to an intronic element within the Stim1 gene. We show that NEUROD2 limits Stim1 expression in cortical neurons and consequently fine-tunes the SOCE response upon depletion of ER calcium. Our findings reveal a novel mechanism that regulates neuronal calcium homeostasis during cortical development.
Collapse
|
22
|
Quednow BB, Brzózka MM, Rossner MJ. Transcription factor 4 (TCF4) and schizophrenia: integrating the animal and the human perspective. Cell Mol Life Sci 2014; 71:2815-35. [PMID: 24413739 PMCID: PMC11113759 DOI: 10.1007/s00018-013-1553-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 12/04/2013] [Accepted: 12/30/2013] [Indexed: 02/06/2023]
Abstract
Schizophrenia is a genetically complex disease considered to have a neurodevelopmental pathogenesis and defined by a broad spectrum of positive and negative symptoms as well as cognitive deficits. Recently, large genome-wide association studies have identified common alleles slightly increasing the risk for schizophrenia. Among the few schizophrenia-risk genes that have been consistently replicated is the basic Helix-Loop-Helix (bHLH) transcription factor 4 (TCF4). Haploinsufficiency of the TCF4 (formatting follows IUPAC nomenclature: TCF4 protein/protein function, Tcf4 rodent gene cDNA mRNA, TCF4 human gene cDNA mRNA) gene causes the Pitt-Hopkins syndrome-a neurodevelopmental disease characterized by severe mental retardation. Accordingly, Tcf4 null-mutant mice display developmental brain defects. TCF4-associated risk alleles are located in putative coding and non-coding regions of the gene. Hence, subtle changes at the level of gene expression might be relevant for the etiopathology of schizophrenia. Behavioural phenotypes obtained with a mouse model of slightly increased gene dosage and electrophysiological investigations with human risk-allele carriers revealed an overlapping spectrum of schizophrenia-relevant endophenotypes. Most prominently, early information processing and higher cognitive functions appear to be associated with TCF4 risk genotypes. Moreover, a recent human study unravelled gene × environment interactions between TCF4 risk alleles and smoking behaviour that were specifically associated with disrupted early information processing. Taken together, TCF4 is considered as an integrator ('hub') of several bHLH networks controlling critical steps of various developmental, and, possibly, plasticity-related transcriptional programs in the CNS and changes of TCF4 expression also appear to affect brain networks important for information processing. Consequently, these findings support the neurodevelopmental hypothesis of schizophrenia and provide a basis for identifying the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Boris B. Quednow
- Department of Psychiatry, Psychotherapy and Psychosomatics, Experimental and Clinical Pharmacopsychology, Psychiatric Hospital, University of Zurich, Lenggstrasse 31, 8032 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Magdalena M. Brzózka
- Department of Psychiatry, Molecular and Behavioral Neurobiology, Ludwig-Maximillians-University, Nussbaumstr. 7, 80336 Munich, Germany
| | - Moritz J. Rossner
- Department of Psychiatry, Molecular and Behavioral Neurobiology, Ludwig-Maximillians-University, Nussbaumstr. 7, 80336 Munich, Germany
- Research Group Gene Expression, Max-Planck-Institute of Experimental Medicine, Hermann-Rein-Str. 3, Goettingen, 37075 Germany
| |
Collapse
|
23
|
Transcriptional analysis of apoptotic cerebellar granule neurons following rescue by gastric inhibitory polypeptide. Int J Mol Sci 2014; 15:5596-622. [PMID: 24694544 PMCID: PMC4013584 DOI: 10.3390/ijms15045596] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/04/2014] [Accepted: 03/17/2014] [Indexed: 12/31/2022] Open
Abstract
Apoptosis triggered by exogenous or endogenous stimuli is a crucial phenomenon to determine the fate of neurons, both in physiological and in pathological conditions. Our previous study established that gastric inhibitory polypeptide (Gip) is a neurotrophic factor capable of preventing apoptosis of cerebellar granule neurons (CGNs), during its pre-commitment phase. In the present study, we conducted whole-genome expression profiling to obtain a comprehensive view of the transcriptional program underlying the rescue effect of Gip in CGNs. By using DNA microarray technology, we identified 65 genes, we named survival related genes, whose expression is significantly de-regulated following Gip treatment. The expression levels of six transcripts were confirmed by real-time quantitative polymerase chain reaction. The proteins encoded by the survival related genes are functionally grouped in the following categories: signal transduction, transcription, cell cycle, chromatin remodeling, cell death, antioxidant activity, ubiquitination, metabolism and cytoskeletal organization. Our data outline that Gip supports CGNs rescue via a molecular framework, orchestrated by a wide spectrum of gene actors, which propagate survival signals and support neuronal viability.
Collapse
|
24
|
Induced neural stem cells: Methods of reprogramming and potential therapeutic applications. Prog Neurobiol 2014; 114:15-24. [DOI: 10.1016/j.pneurobio.2013.11.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 10/18/2013] [Accepted: 11/07/2013] [Indexed: 01/10/2023]
|
25
|
Bormuth I, Yan K, Yonemasu T, Gummert M, Zhang M, Wichert S, Grishina O, Pieper A, Zhang W, Goebbels S, Tarabykin V, Nave KA, Schwab MH. Neuronal basic helix-loop-helix proteins Neurod2/6 regulate cortical commissure formation before midline interactions. J Neurosci 2013; 33:641-51. [PMID: 23303943 PMCID: PMC6704922 DOI: 10.1523/jneurosci.0899-12.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 10/29/2012] [Accepted: 11/05/2012] [Indexed: 12/31/2022] Open
Abstract
Establishment of long-range fiber tracts by neocortical projection neurons is fundamental for higher brain functions. The molecular control of axon tract formation, however, is still poorly understood. Here, we have identified basic helix-loop-helix (bHLH) transcription factors Neurod2 and Neurod6 as key regulators of fasciculation and targeted axogenesis in the mouse neocortex. In Neurod2/6 double-mutant mice, callosal axons lack expression of the cell adhesion molecule Contactin2, defasciculate in the subventricular zone, and fail to grow toward the midline without forming Probst bundles. Instead, mutant axons overexpress Robo1 and follow random trajectories into the ipsilateral cortex. In contrast to long-range axogenesis, generation and maintenance of pyramidal neurons and initial axon outgrowth are grossly normal, suggesting that these processes are under distinct transcriptional control. Our findings define a new stage in corpus callosum development and demonstrate that neocortical projection neurons require transcriptional specification by neuronal bHLH proteins to execute an intrinsic program of remote connectivity.
Collapse
Affiliation(s)
- Ingo Bormuth
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, D-37075 Göttingen, Germany
- Charité–Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, NeuroCure Cluster of Excellence, D-10115 Berlin, Germany, and
| | - Kuo Yan
- Charité–Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, NeuroCure Cluster of Excellence, D-10115 Berlin, Germany, and
| | - Tomoko Yonemasu
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, D-37075 Göttingen, Germany
| | - Maike Gummert
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, D-37075 Göttingen, Germany
| | - Mingyue Zhang
- University of Münster, Department of Psychiatry, Laboratory of Molecular Psychiatry, D-48149 Münster, Germany
| | - Sven Wichert
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, D-37075 Göttingen, Germany
| | - Olga Grishina
- Charité–Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, NeuroCure Cluster of Excellence, D-10115 Berlin, Germany, and
| | - Alexander Pieper
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, D-37075 Göttingen, Germany
| | - Weiqi Zhang
- University of Münster, Department of Psychiatry, Laboratory of Molecular Psychiatry, D-48149 Münster, Germany
| | - Sandra Goebbels
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, D-37075 Göttingen, Germany
| | - Victor Tarabykin
- Charité–Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, NeuroCure Cluster of Excellence, D-10115 Berlin, Germany, and
| | - Klaus-Armin Nave
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, D-37075 Göttingen, Germany
| | - Markus H. Schwab
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, D-37075 Göttingen, Germany
| |
Collapse
|
26
|
Nishioka M, Bundo M, Koike S, Takizawa R, Kakiuchi C, Araki T, Kasai K, Iwamoto K. Comprehensive DNA methylation analysis of peripheral blood cells derived from patients with first-episode schizophrenia. J Hum Genet 2012; 58:91-7. [PMID: 23235336 DOI: 10.1038/jhg.2012.140] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epidemiological studies have revealed that schizophrenia is highly heritable. However, genetic studies have not fully elucidated its etiology. Accumulating evidence suggests that epigenetic alterations may provide an additional explanation of its pathophysiology. We investigated the methylation profiles of DNA in peripheral blood cells from 18 patients with first-episode schizophrenia (FESZ) and from 15 normal controls. Schizophrenia patients were confined to those at the stage of first-episode psychosis. We analyzed the DNA methylation status of 27,578 CpG sites by means of the Illumina Infinium HumanMethylation27 BeadChip array. Differentially methylated CpG sites, which were particularly abundant within CpG islands, were enriched in genes related to the nuclear lumen, to transcription factor binding, and to nucleotide binding. We also observed differential methylation of the promoters of HTR1E and COMTD1, which are functionally related to genes found to be differentially methylated in schizophrenia patients in previous studies. Our results indicate the site-specific epigenetic alterations in patients with FESZ.
Collapse
Affiliation(s)
- Masaki Nishioka
- Department of Molecular Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Messmer K, Shen WB, Remington M, Fishman PS. Induction of neural differentiation by the transcription factor neuroD2. Int J Dev Neurosci 2011; 30:105-12. [PMID: 22197973 DOI: 10.1016/j.ijdevneu.2011.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 09/30/2011] [Accepted: 12/09/2011] [Indexed: 01/08/2023] Open
Abstract
Pro-neural basic helix loop helix (bHLH) transcription factors are involved in many aspects of normal neuronal development, and over-expression of genes for several of these factors has been shown to induce aspects of neuronal differentiation in cell lines and stem cells. Here we show that over-expression of NeuroD2 (ND2), Neurogenin1 and 2 leads to morphological differentiation of N18-RE-105 neuroblastoma cells and increased expression of synaptic proteins. Particularly ND2 induced neurite formation and increases in the expression of synaptic proteins such as synaptotagmin, that is not expressed normally in this cell type, as well as the redistribution of another synaptic protein, SNAP25, to a cell membrane location. Infection of human neural progenitor cells using adeno associated viral (AAV) vectors also promoted neuronal differentiation. Over-expressing cells demonstrated a significant increase in the neuron specific form of tubulin as well as increased expression of synaptotagmin. Genetic modification of neural progenitor cell with bHLH factors such as ND2 may be a viable strategy to enhance differentiation of these cells into replacement neurons for human disease.
Collapse
Affiliation(s)
- Kirsten Messmer
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
28
|
MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 2011; 476:228-31. [PMID: 21753754 DOI: 10.1038/nature10323] [Citation(s) in RCA: 754] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 06/27/2011] [Indexed: 12/13/2022]
Abstract
Neurogenic transcription factors and evolutionarily conserved signalling pathways have been found to be instrumental in the formation of neurons. However, the instructive role of microRNAs (miRNAs) in neurogenesis remains unexplored. We recently discovered that miR-9* and miR-124 instruct compositional changes of SWI/SNF-like BAF chromatin-remodelling complexes, a process important for neuronal differentiation and function. Nearing mitotic exit of neural progenitors, miR-9* and miR-124 repress the BAF53a subunit of the neural-progenitor (np)BAF chromatin-remodelling complex. After mitotic exit, BAF53a is replaced by BAF53b, and BAF45a by BAF45b and BAF45c, which are then incorporated into neuron-specific (n)BAF complexes essential for post-mitotic functions. Because miR-9/9* and miR-124 also control multiple genes regulating neuronal differentiation and function, we proposed that these miRNAs might contribute to neuronal fates. Here we show that expression of miR-9/9* and miR-124 (miR-9/9*-124) in human fibroblasts induces their conversion into neurons, a process facilitated by NEUROD2. Further addition of neurogenic transcription factors ASCL1 and MYT1L enhances the rate of conversion and the maturation of the converted neurons, whereas expression of these transcription factors alone without miR-9/9*-124 was ineffective. These studies indicate that the genetic circuitry involving miR-9/9*-124 can have an instructive role in neural fate determination.
Collapse
|
29
|
Zill P, Preuss UW, Koller G, Bondy B, Soyka M. Lack of association between SNPs in the NEUROD2 gene and alcohol dependence in a German patient sample. Psychiatry Res 2011; 187:220-3. [PMID: 20880594 DOI: 10.1016/j.psychres.2010.08.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 08/20/2010] [Accepted: 08/29/2010] [Indexed: 10/19/2022]
Abstract
Results of a human post mortem study performed by our own group have suggested that the transcription factor NEUROD2, which plays a role in neuronal development, as well as in the development of anxiety and risk behavior in mice, might be a susceptibility factor for addictive disorders. Therefore the aim of the present study was to analyze a possible relation between genetic variants in the NEUROD2 gene and alcohol dependence in a sample of the Munich Gene Bank of Alcoholism (MGBA). We performed single SNP (single nucleotide polymorphism) and haplotype studies in 430 alcohol-dependent patients and 365 healthy controls with four SNPs covering the gene region of NEUROD2. Neither single SNP nor haplotype analysis could detect significant associations with alcohol dependence. Additionally we could not detect any relation of the analyzed genetic variants to Cloninger's Type 1/2 or Babor's Type A/B classification, to the age of onset or to the amount of alcohol intake. Our results do not provide evidence for an involvement of NEUROD2 polymorphisms in the pathophysiology of alcohol dependence. Further association studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Peter Zill
- Department of Psychiatry, Ludwig-Maximilians-University, Munich, Germany.
| | | | | | | | | |
Collapse
|
30
|
Cognitive and sensorimotor gating impairments in transgenic mice overexpressing the schizophrenia susceptibility gene Tcf4 in the brain. Biol Psychiatry 2010; 68:33-40. [PMID: 20434134 DOI: 10.1016/j.biopsych.2010.03.015] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 02/10/2010] [Accepted: 03/03/2010] [Indexed: 01/25/2023]
Abstract
BACKGROUND The combined analysis of several large genome-wide association studies identified the basic helix-loop-helix (bHLH) transcription factor TCF4 as one of the most significant schizophrenia susceptibility genes. Its function in the adult brain, however, is not known. TCF4 belongs to the E-protein subfamily known to be involved in neurodevelopment. The messenger RNA expression of Tcf4 is sustained in the adult mouse brain, suggesting a function in the adult nervous system. Tcf4 null mutant mice die perinatally, and haploinsufficiency of TCF4 in humans causes severe mental retardation. METHODS To investigate the possible function of TCF4 in the adult central nervous system, we generated transgenic mice that moderately overexpress TCF4 postnatally in the brain to reduce the risk of developmental effects possibly interfering with adult brain functions. Tcf4 transgenic mice were characterized with molecular, histological, and behavioral methods. RESULTS Tcf4 transgenic mice display profound deficits in contextual and cued fear conditioning and sensorimotor gating. Furthermore, we show that TCF4 interacts with the neurogenic bHLH factors NEUROD and NDRF in vivo. Molecular analyses revealed the dynamic circadian deregulation of neuronal bHLH factors in the adult hippocampus. CONCLUSIONS We conclude that TCF4 likely acts in concert with other neuronal bHLH transcription factors contributing to higher-order cognitive processing. Moderate transcriptional deregulation of Tcf4 in the brain interferes with cognitive functions and might alter circadian processes in mice. These observations provide insight for the first time into the physiological function of TCF4 in the adult brain and its possible contributions to neuropsychiatric disease conditions.
Collapse
|
31
|
Ravanpay AC, Hansen SJ, Olson JM. Transcriptional inhibition of REST by NeuroD2 during neuronal differentiation. Mol Cell Neurosci 2010; 44:178-89. [PMID: 20346398 DOI: 10.1016/j.mcn.2010.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 03/07/2010] [Accepted: 03/08/2010] [Indexed: 11/17/2022] Open
Abstract
For a progenitor cell to become a neuron, three activities must occur: neuronal differentiation program must be activated, elements repressing neuronal differentiation must be deactivated and competing differentiation programs must be silenced. It is known that NeuroD2 and related bHLH transcription factors induce neuronal differentiation, REST represses neuronal differentiation, and Zfhx1a prevents myogenic gene expression. We demonstrate that NeuroD2 suppresses REST during differentiation in culture. In the hippocampus of NeuroD2 knockout mice, higher level of REST is detected. Functional significance of NeuroD2-REST interplay is uncovered by showing that forced expression of REST interferes with neuronal differentiation in culture. NeuroD2 inhibits REST indirectly by involving the inhibitor of myogenic genes, Zfhx1a, which binds response elements in REST 5'-UTR. Our study supports a model wherein NeuroD2 induces transcription of neuronal genes and Zfhx1a, which in turn de-represses neuronal differentiation by down-regulating REST, and suppresses competing myogenic fate.
Collapse
Affiliation(s)
- Ali C Ravanpay
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | |
Collapse
|
32
|
Yang Y, Kim AH, Yamada T, Wu B, Bilimoria PM, Ikeuchi Y, de la Iglesia N, Shen J, Bonni A. A Cdc20-APC ubiquitin signaling pathway regulates presynaptic differentiation. Science 2009; 326:575-8. [PMID: 19900895 DOI: 10.1126/science.1177087] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Presynaptic axonal differentiation is essential for synapse formation and the establishment of neuronal circuits. However, the mechanisms that coordinate presynaptic development in the brain are largely unknown. We found that the major mitotic E3 ubiquitin ligase Cdc20-anaphase promoting complex (Cdc20-APC) regulates presynaptic differentiation in primary postmitotic mammalian neurons and in the rat cerebellar cortex. Cdc20-APC triggered the degradation of the transcription factor NeuroD2 and thereby promoted presynaptic differentiation. The NeuroD2 target gene encoding Complexin II, which acts locally at presynaptic sites, mediated the ability of NeuroD2 to suppress presynaptic differentiation. Thus, our findings define a Cdc20-APC ubiquitin signaling pathway that governs presynaptic development, which holds important implications for neuronal connectivity and plasticity in the brain.
Collapse
Affiliation(s)
- Yue Yang
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Yang RJ, Mozhui K, Karlsson RM, Cameron HA, Williams RW, Holmes A. Variation in mouse basolateral amygdala volume is associated with differences in stress reactivity and fear learning. Neuropsychopharmacology 2008; 33:2595-604. [PMID: 18185497 DOI: 10.1038/sj.npp.1301665] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A wealth of research identifies the amygdala as a key brain region mediating negative affect, and implicates amygdala dysfunction in the pathophysiology of anxiety disorders. Although there is a strong genetic component to anxiety disorders such as posttraumatic stress disorder (PTSD) there remains debate about whether abnormalities in amygdala function predispose to these disorders. In the present study, groups of C57BL/6 x DBA/2 (B x D) recombinant inbred strains of mice were selected for differences in volume of the basolateral amygdala complex (BLA). Strains with relatively small, medium, or large BLA volumes were compared for Pavlovian fear learning and memory, anxiety-related behaviors, depression-related behavior, and glucocorticoid responses to stress. Strains with relatively small BLA exhibited stronger conditioned fear responses to both auditory tone and contextual stimuli, as compared to groups with larger BLA. The small BLA group also showed significantly greater corticosterone responses to stress than the larger BLA groups. BLA volume did not predict clear differences in measures of anxiety-like behavior or depression-related behavior, other than greater locomotor inhibition to novelty in strains with smaller BLA. Neither striatal, hippocampal nor cerebellar volumes correlated significantly with any behavioral measure. The present data demonstrate a phenotype of enhanced fear conditioning and exaggerated glucocorticoid responses to stress associated with small BLA volume. This profile is reminiscent of the increased fear processing and stress reactivity that is associated with amygdala excitability and reduced amygdala volume in humans carrying loss of function polymorphisms in the serotonin transporter and monoamine oxidase A genes. Our study provides a unique example of how natural variation in amygdala volume associates with specific fear- and stress-related phenotypes in rodents, and further supports the role of amygdala dysfunction in anxiety disorders such as PTSD.
Collapse
Affiliation(s)
- Rebecca J Yang
- Section on Behavioral Science and Genetics, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Rockville, MD 20852-9411, USA
| | | | | | | | | | | |
Collapse
|
34
|
Ravanpay AC, Olson JM. E protein dosage influences brain development more than family member identity. J Neurosci Res 2008; 86:1472-81. [PMID: 18214987 DOI: 10.1002/jnr.21615] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Loss-of-function studies have revealed the role of many basic helix-loop-helix (bHLH) transcription factors at specific points during development; however, the role of E proteins in the development of the nervous system has not been experimentally addressed. E proteins have been speculated to interact selectively with class II bHLH factors to form different neurogenic complexes. In this study, using coimmunoprecipitation in a culture model of neurogenesis (P19 cells), we show that E proteins E12, HEB, and E2-2 interact with neuroD2. Using electrophoretic mobility shift assay and P19 cell culture, we show that these heterodimers bind a neuroD2 preferred E box and induce neurogenesis equally well. We examine the mRNA levels of the three E proteins at 10 time points during brain development and show that E protein gene expression is regulated such that at certain times during development selective interaction between neuroD2 and a single E protein (HEB) is a possibility. This led us to study the brains of HEB and E2A knockout mice, which manifest no gross neuroanatomical, cellular, or behavioral deficits. These findings, together with homology in the primary peptide sequence of E proteins, suggest functional compensation among E proteins during development of the nervous system.
Collapse
Affiliation(s)
- Ali C Ravanpay
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | |
Collapse
|
35
|
Cudilo E, Al Naemi H, Marmorstein L, Baldwin AL. Knockout mice: is it just genetics? Effect of enriched housing on fibulin-4(+/-) mice. PLoS One 2007; 2:e229. [PMID: 17311093 PMCID: PMC1794134 DOI: 10.1371/journal.pone.0000229] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2006] [Accepted: 01/28/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Fibulin-4 is an extracellular matrix protein expressed by vascular smooth muscle cells that is essential for maintaining arterial integrity. Fibulin-4(-/-) mice die just before birth due to arterial hemorrhage, but fibulin-4(+/-) mice appear to be outwardly normal. Experiments were therefore performed to determine whether fibulin-4(+/-) mice display arterial pathologies on a microscopic scale. After preliminary experiments were performed, a second purpose developed, which was to test the hypothesis that any observed pathologies would be ameliorated by housing the animals in enriched cages. METHODOLOGY Fibulin-4(+/-) and wild-type mice were housed either four/cage in standard cages or two per cage in larger cages, each cage containing a tunnel and a wheel. After three weeks the mice were sacrificed, and the aortas perfusion-fixed and excised for light and electron microscopy. PRINCIPLE FINDINGS When the mice were in standard cages, localized regions of disorganized extracellular matrix and collagen fibers consistently appeared between some of the medial smooth muscle cells in the fibulin-4(+/-) mice. In the wild-type mice, the smooth muscle cells were closely connected to each other and the media was more compact. The number of disorganized regions per square mm was significantly greater for fibulin-4(+/-) mice (172+/-43 (SEM)) than for wild-type mice (15+/-8) (p<0.01, n = 8). When the mice were in enriched cages, the fibulin-4(+/-) mice showed significantly fewer disorganized regions than those in standard cages (35+/-12) (p<0.05, n = 8). The wild type mice also showed fewer disorganized regions (3+/-2), but this difference was not significant. CONCLUSIONS These results indicate that arterial pathologies manifested in fibulin-4(+/-) mice can be reduced by enriching the housing conditions, and imply that appropriate environments may counteract the effects of some genetic deficiencies.
Collapse
MESH Headings
- Animals
- Aorta/abnormalities
- Aorta/growth & development
- Aorta/ultrastructure
- Crosses, Genetic
- Endothelium, Vascular/abnormalities
- Endothelium, Vascular/ultrastructure
- Environment Design
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/physiology
- Fibrosis
- Heterozygote
- Housing, Animal
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout/genetics
- Motor Activity
- Muscle, Smooth, Vascular/abnormalities
- Muscle, Smooth, Vascular/growth & development
- Physical Conditioning, Animal
- Research Design
- Tight Junctions/ultrastructure
Collapse
Affiliation(s)
- Elizabeth Cudilo
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona, United States of America
| | | | - Lihua Marmorstein
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona, United States of America
| | - Ann L. Baldwin
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
36
|
Mozhui K, Hamre KM, Holmes A, Lu L, Williams RW. Genetic and Structural Analysis of the Basolateral Amygdala Complex in BXD Recombinant Inbred Mice. Behav Genet 2006; 37:223-43. [PMID: 17131200 DOI: 10.1007/s10519-006-9122-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Accepted: 10/13/2006] [Indexed: 12/21/2022]
Abstract
The amygdala integrates and coordinates emotional and autonomic responses. The genetics that underlie variation in amygdala structure may be coupled to variation in levels of aggression, fear, anxiety, and affiliated behaviors. We systematically quantified the volume and cell populations of the basolateral amygdala complex (BLAc) across 35 BXD recombinant inbred (RI) lines, the parental strains--C57BL/6J (B6) and DBA/2J (D2)--and F1 hybrids (n cases=199, bilateral analysis). Neuron number and volume vary 1.7- to 2-fold among strains (e.g., neuron number ranged from 88,000 to 170,000). Glial and endothelial populations ranged more widely (5- to 8-fold), in part because of higher technical error. A quantitative trait locus (QTL) for the BLAc size is located on chromosome (Chr) 8 near the Large gene. This locus may also influence volume of other regions including hippocampus and cerebellum. Cell populations in the BLAc appear to be modulated more weakly by loci on Chrs 11 and 13. Candidate genes were selected on the basis of correlation with BLAc traits, chromosomal location, single nucleotide polymorphism (SNP) density, and expression patterns in the Allen Brain Atlas. Neurod2, a gene shown to be significant for the formation of the BLAc by knockout studies, is among the candidates genes. Other candidates include Large, and Thra. Responses to drugs of abuse and locomotor activity were the most notable behavioral correlates of the BLAc traits.
Collapse
Affiliation(s)
- Khyobeni Mozhui
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, 855 Monroe Avenue, Memphis, TN 38163, USA
| | | | | | | | | |
Collapse
|
37
|
Till BJ, Zerr T, Bowers E, Greene EA, Comai L, Henikoff S. High-throughput discovery of rare human nucleotide polymorphisms by Ecotilling. Nucleic Acids Res 2006; 34:e99. [PMID: 16893952 PMCID: PMC1540726 DOI: 10.1093/nar/gkl479] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human individuals differ from one another at only approximately 0.1% of nucleotide positions, but these single nucleotide differences account for most heritable phenotypic variation. Large-scale efforts to discover and genotype human variation have been limited to common polymorphisms. However, these efforts overlook rare nucleotide changes that may contribute to phenotypic diversity and genetic disorders, including cancer. Thus, there is an increasing need for high-throughput methods to robustly detect rare nucleotide differences. Toward this end, we have adapted the mismatch discovery method known as Ecotilling for the discovery of human single nucleotide polymorphisms. To increase throughput and reduce costs, we developed a universal primer strategy and implemented algorithms for automated band detection. Ecotilling was validated by screening 90 human DNA samples for nucleotide changes in 5 gene targets and by comparing results to public resequencing data. To increase throughput for discovery of rare alleles, we pooled samples 8-fold and found Ecotilling to be efficient relative to resequencing, with a false negative rate of 5% and a false discovery rate of 4%. We identified 28 new rare alleles, including some that are predicted to damage protein function. The detection of rare damaging mutations has implications for models of human disease.
Collapse
Affiliation(s)
- Bradley J Till
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Lin CH, Tapscott SJ, Olson JM. Congenital hypothyroidism (cretinism) in neuroD2-deficient mice. Mol Cell Biol 2006; 26:4311-5. [PMID: 16705180 PMCID: PMC1489089 DOI: 10.1128/mcb.02158-05] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mice lacking neuroD2, a basic helix-loop-helix transcription factor involved in brain development, show growth retardation and other abnormalities consistent with hypothalamic-pituitary-thyroid (HPT) axis dysfunction. neuroD2 is expressed in the paraventricular hypothalamic nuclei, the anterior lobe of pituitary, and the thyroid gland. In neuroD2-deficient mice, thyrotropin-releasing hormone, thyroid-stimulating hormone, and thyroid hormone are decreased in these three regions, respectively. neuroD2-null mice typically die 2 to 3 weeks after birth, but those treated with replacement doses of thyroxine survived more than 8 weeks. These data indicate that neuroD2 is expressed throughout the HPT axis and that all levels of the axis are functionally affected by its absence in mice.
Collapse
Affiliation(s)
- Chin-Hsing Lin
- Clinical Research, Fred Hutchinson Cancer Research Center, Mailstop D4-100, 1100 Fairview Ave. N., Seattle, WA 98109, USA
| | | | | |
Collapse
|
39
|
Zill P, Büttner A, Eisenmenger W, Bondy B. A possible impact of the neuroD2 transcription factor on the development of drug abusing behavior. Mol Psychiatry 2006; 11:525-7. [PMID: 16607387 DOI: 10.1038/sj.mp.4001825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|