137 results on '"Gail Mandel"'
Search Results
2. In Vivo Repair of a Protein Underlying a Neurological Disorder by Programmable RNA Editing
- Author
-
John R. Sinnamon, Susan Y. Kim, Jenna R. Fisk, Zhen Song, Hiroyuki Nakai, Sophia Jeng, Shannon K. McWeeney, and Gail Mandel
- Subjects
MeCP2 ,Rett syndrome ,adenosine deaminase acting on RNA ,ADAR ,RNA editing ,Biology (General) ,QH301-705.5 - Abstract
Summary: Programmable RNA editing is gaining momentum as an approach to repair mutations, but its efficiency in repairing endogenous mutant RNA in complex tissue is unknown. Here we apply this approach to the brain and successfully repair a guanosine-to-adenosine mutation in methyl CpG binding protein 2 RNA that causes the neurodevelopmental disease Rett syndrome. Repair is mediated by hippocampal injections of juvenile Mecp2317G>A mice with an adeno-associated virus expressing the hyperactive catalytic domain of adenosine deaminase acting on RNA 2 and Mecp2 guide. After 1 month, 50% of Mecp2 RNA is recoded in three different hippocampal neuronal populations. MeCP2 protein localization to heterochromatin is restored in neurons to 50% of wild-type levels. Whole-transcriptome RNA analysis of one neuronal population indicates that the majority of off-target editing sites exhibit rates of 30% or less. This study demonstrates that programmable RNA editing can be utilized to repair mutations in mouse models of neurological disease.
- Published
- 2020
- Full Text
- View/download PDF
3. Astrocytic modulation of excitatory synaptic signaling in a mouse model of Rett syndrome
- Author
-
Benjamin Rakela, Paul Brehm, and Gail Mandel
- Subjects
Neuroscience ,astrocytes ,astrocyte-neuron signaling ,Rett syndrome ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Studies linking mutations in Methyl CpG Binding Protein 2 (MeCP2) to physiological defects in the neurological disease, Rett syndrome, have focused largely upon neuronal dysfunction despite MeCP2 ubiquitous expression. Here we explore roles for astrocytes in neuronal network function using cortical slice recordings. We find that astrocyte stimulation in wild-type mice increases excitatory synaptic activity that is absent in male mice lacking MeCP2 globally. To determine the cellular basis of the defect, we exploit a female mouse model for Rett syndrome that expresses wild-type MeCP2-GFP in a mosaic distribution throughout the brain, allowing us to test all combinations of wild-type and mutant cells. We find that the defect is dependent upon MeCP2 expression status in the astrocytes and not in the neurons. Our findings highlight a new role for astrocytes in regulation of excitatory synaptic signaling and in the neurological defects associated with Rett syndrome.
- Published
- 2018
- Full Text
- View/download PDF
4. The REST remodeling complex protects genomic integrity during embryonic neurogenesis
- Author
-
Tamilla Nechiporuk, James McGann, Karin Mullendorff, Jenny Hsieh, Wolfgang Wurst, Thomas Floss, and Gail Mandel
- Subjects
transcription factor ,repression ,genomic instability ,neurogenesis ,REST complex ,knockout animal ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The timely transition from neural progenitor to post-mitotic neuron requires down-regulation and loss of the neuronal transcriptional repressor, REST. Here, we have used mice containing a gene trap in the Rest gene, eliminating transcription from all coding exons, to remove REST prematurely from neural progenitors. We find that catastrophic DNA damage occurs during S-phase of the cell cycle, with long-term consequences including abnormal chromosome separation, apoptosis, and smaller brains. Persistent effects are evident by latent appearance of proneural glioblastoma in adult mice deleted additionally for the tumor suppressor p53 protein (p53). A previous line of mice deleted for REST in progenitors by conventional gene targeting does not exhibit these phenotypes, likely due to a remaining C-terminal peptide that still binds chromatin and recruits co-repressors. Our results suggest that REST-mediated chromatin remodeling is required in neural progenitors for proper S-phase dynamics, as part of its well-established role in repressing neuronal genes until terminal differentiation.
- Published
- 2016
- Full Text
- View/download PDF
5. Polycomb- and REST-associated histone deacetylases are independent pathways toward a mature neuronal phenotype
- Author
-
James C McGann, Jon A Oyer, Saurabh Garg, Huilan Yao, Jun Liu, Xin Feng, Lujian Liao, John R Yates III, and Gail Mandel
- Subjects
ES cell ,REST ,Polycomb ,poised ,histone deacetylase ,neuronal ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The bivalent hypothesis posits that genes encoding developmental regulators required for early lineage decisions are poised in stem/progenitor cells by the balance between a repressor histone modification (H3K27me3), mediated by the Polycomb Repressor Complex 2 (PRC2), and an activator modification (H3K4me3). In this study, we test whether this mechanism applies equally to genes that are not required until terminal differentiation. We focus on the RE1 Silencing Transcription Factor (REST) because it is expressed highly in stem cells and is an established global repressor of terminal neuronal genes. Elucidation of the REST complex, and comparison of chromatin marks and gene expression levels in control and REST-deficient stem cells, shows that REST target genes are poised by a mechanism independent of Polycomb, even at promoters which bear the H3K27me3 mark. Specifically, genes under REST control are actively repressed in stem cells by a balance of the H3K4me3 mark and a repressor complex that relies on histone deacetylase activity. Thus, chromatin distinctions between pro-neural and terminal neuronal genes are established at the embryonic stem cell stage by two parallel, but distinct, repressor pathways.
- Published
- 2014
- Full Text
- View/download PDF
6. Synchronous and asynchronous modes of synaptic transmission utilize different calcium sources
- Author
-
Hua Wen, Jeffrey M Hubbard, Benjamin Rakela, Michael W Linhoff, Gail Mandel, and Paul Brehm
- Subjects
synaptopHluorin ,calcium indicator ,motor neuron ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Asynchronous transmission plays a prominent role at certain synapses but lacks the mechanistic insights of its synchronous counterpart. The current view posits that triggering of asynchronous release during repetitive stimulation involves expansion of the same calcium domains underlying synchronous transmission. In this study, live imaging and paired patch clamp recording at the zebrafish neuromuscular synapse reveal contributions by spatially distinct calcium sources. Synchronous release is tied to calcium entry into synaptic boutons via P/Q type calcium channels, whereas asynchronous release is boosted by a propagating intracellular calcium source initiated at off-synaptic locations in the axon and axonal branch points. This secondary calcium source fully accounts for the persistence following termination of the stimulus and sensitivity to slow calcium buffers reported for asynchronous release. The neuromuscular junction and CNS neurons share these features, raising the possibility that secondary calcium sources are common among synapses with prominent asynchronous release.
- Published
- 2013
- Full Text
- View/download PDF
7. Library Screening Reveals Sequence Motifs That Enable ADAR2 Editing at Recalcitrant Sites
- Author
-
Casey S. Jacobsen, Prince Salvador, John F. Yung, Sabrina Kragness, Herra G. Mendoza, Gail Mandel, and Peter A. Beal
- Subjects
Molecular Medicine ,General Medicine ,Biochemistry - Published
- 2023
8. Editor's evaluation: Bidirectional regulation of postmitotic H3K27me3 distributions underlie cerebellar granule neuron maturation dynamics
- Author
-
Gail Mandel
- Published
- 2023
9. Targeted RNA editing in brainstem alleviates respiratory dysfunction in a mouse model of Rett syndrome
- Author
-
John R. Sinnamon, Michael E. Jacobson, John F. Yung, Jenna R. Fisk, Sophia Jeng, Shannon K. McWeeney, Lindsay K. Parmelee, Chi Ngai Chan, Siu-Pok Yee, and Gail Mandel
- Subjects
Male ,Disease Models, Animal ,Mice ,Multidisciplinary ,Methyl-CpG-Binding Protein 2 ,Mutation ,Rett Syndrome ,Animals ,Humans ,RNA Editing ,Respiration Disorders ,Brain Stem - Abstract
Rett syndrome is a neurological disease due to loss-of-function mutations in the transcription factor, Methyl CpG binding protein 2 (MECP2). Because overexpression of endogenous MECP2 also causes disease, we have exploited a targeted RNA-editing approach to repair patient mutations where levels of MECP2 protein will never exceed endogenous levels. Here, we have constructed adeno-associated viruses coexpressing a bioengineered wild-type ADAR2 catalytic domain (Editase wt ) and either Mecp2 -targeting or nontargeting gfp RNA guides. The viruses are introduced systemically into male mice containing a guanosine to adenosine mutation that eliminates MeCP2 protein and causes classic Rett syndrome in humans. We find that in the mutant mice injected with the Mecp2 -targeting virus, the brainstem exhibits the highest RNA-editing frequency compared to other brain regions. The efficiency is sufficient to rescue MeCP2 expression and function in the brainstem of mice expressing the Mecp2 -targeting virus. Correspondingly, we find that abnormal Rett-like respiratory patterns are alleviated, and survival is prolonged, compared to mice injected with the control gfp guide virus. The levels of RNA editing among most brain regions corresponds to the distribution of guide RNA rather than Editase wt . Our results provide evidence that a targeted RNA-editing approach can alleviate a hallmark symptom in a mouse model of human disease.
- Published
- 2023
10. The Genome-Wide Binding Profile for Human RE1 Silencing Transcription Factor Unveils a Unique Genetic Circuitry in Hippocampus
- Author
-
Randall L. Woltjer, James C. McGann, Karin Mullendorff, Gail Mandel, Michael A. Spinner, and Saurabh K. Garg
- Subjects
Male ,Aging ,Cell type ,Repressor ,RE1-silencing transcription factor ,Hippocampus ,Mice ,Gene expression ,medicine ,Animals ,Humans ,Gene ,Research Articles ,Aged ,Neurons ,Regulation of gene expression ,biology ,General Neuroscience ,Neurogenesis ,Human brain ,Middle Aged ,Immunity, Innate ,Cell biology ,Repressor Proteins ,medicine.anatomical_structure ,biology.protein ,Female ,Neuroglia ,Genome-Wide Association Study - Abstract
Early studies in mouse neurodevelopment led to the discovery of the RE1 Silencing Transcription Factor (REST) and its role as a master repressor of neuronal gene expression. Recently, REST was reported to also repress neuronal genes in the human adult brain. These genes were found to be involved in pro-apoptotic pathways; and their repression, associated with increased REST levels during aging, were found to be neuroprotective and conserved across species. However, direct genome-wide REST binding profiles for REST in adult brain have not been identified for any species. Here, we apply this approach to mouse and human hippocampus. We find an expansion of REST binding sites in the human hippocampus that are lacking in both mouse hippocampus and other human non-neuronal cell types. The unique human REST binding sites are associated with genes involved in innate immunity processes and inflammation signaling which, on the basis of histology and recent public transcriptomic analyses, suggest that these new target genes are repressed in glia. We propose that the increases in REST expression in mid-adulthood presage the beginning of brain aging, and that human REST function has evolved to protect the longevity and function of both neurons and glia in human brain. SIGNIFICANCE STATEMENT The RE1 Silencing Transcription Factor (REST) repressor has served historically as a model for gene regulation during mouse neurogenesis. Recent studies of REST have also suggested a conserved role for REST repressor function across lower species during aging. However, direct genome-wide studies for REST have been lacking for human brain. Here, we perform the first genome-wide analysis of REST binding in both human and mouse hippocampus. The majority of REST-occupied genes in human hippocampus are distinct from those in mouse. Further, the REST-associated genes unique to human hippocampus represent a new set related to innate immunity and inflammation, where their gene dysregulation has been implicated in aging-related neuropathology, such as Alzheimer's disease.
- Published
- 2021
11. Decision letter: Deleting Mecp2 from the cerebellum rather than its neuronal subtypes causes a delay in motor learning in mice
- Author
-
Gail Mandel
- Subjects
Cerebellum ,medicine.anatomical_structure ,medicine ,Biology ,Motor learning ,Neuroscience ,MECP2 - Published
- 2020
12. In Vivo Repair of a Protein Underlying a Neurological Disorder by Programmable RNA Editing
- Author
-
Jenna R. Fisk, Zhen Song, Shannon K. McWeeney, Sophia Jeng, Susan Y. Kim, Gail Mandel, John R. Sinnamon, and Hiroyuki Nakai
- Subjects
0301 basic medicine ,Male ,RNA editing ,Heterochromatin ,Methyl-CpG-Binding Protein 2 ,Rett syndrome ,Biology ,medicine.disease_cause ,Hippocampus ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Article ,MECP2 ,Cell Line ,Stereotaxic Techniques ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Humans ,Amino Acid Sequence ,lcsh:QH301-705.5 ,MeCP2 ,Mutation ,adenosine deaminase acting on RNA ,Gene Expression Profiling ,RNA ,Genetic Therapy ,medicine.disease ,ADAR ,Cell biology ,030104 developmental biology ,HEK293 Cells ,lcsh:Biology (General) ,Nervous System Diseases ,030217 neurology & neurosurgery - Abstract
SUMMARY Programmable RNA editing is gaining momentum as an approach to repair mutations, but its efficiency in repairing endogenous mutant RNA in complex tissue is unknown. Here we apply this approach to the brain and successfully repair a guanosine-to-adenosine mutation in methyl CpG binding protein 2 RNA that causes the neurodevelopmental disease Rett syndrome. Repair is mediated by hippocampal injections of juvenile Mecp2317G>A mice with an adeno-associated virus expressing the hyperactive catalytic domain of adenosine deaminase acting on RNA 2 and Mecp2 guide. After 1 month, 50% of Mecp2 RNA is recoded in three different hippocampal neuronal populations. MeCP2 protein localization to heterochromatin is restored in neurons to 50% of wild-type levels. Whole-transcriptome RNA analysis of one neuronal population indicates that the majority of off-target editing sites exhibit rates of 30% or less. This study demonstrates that programmable RNA editing can be utilized to repair mutations in mouse models of neurological disease., Graphical Abstract, In Brief Sinnamon et al. show that the Mecp2 guide-targeted “editase,” introduced by an adeno-associated virus into the hippocampus of a Rett syndrome mouse model, repairs a large fraction of pathological Mecp2G>A RNA. MeCP2 function, visualized by chromatin association in neurons, is repaired to similar levels. The results point toward a potential therapy for Rett syndrome.
- Published
- 2020
13. In vivo repair of a protein underlying a neurological disorder by programmable RNA editing
- Author
-
Susan Y. Kim, Gail Mandel, John R. Sinnamon, Hiroyuki Nakai, Jenna R. Fisk, Sophia Jeng, Zhen Song, and Shannon K. McWeeney
- Subjects
0303 health sciences ,Mutation ,biology ,Heterochromatin ,RNA ,Rett syndrome ,medicine.disease ,medicine.disease_cause ,Adenosine ,MECP2 ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Adenosine deaminase ,RNA editing ,medicine ,biology.protein ,030217 neurology & neurosurgery ,030304 developmental biology ,medicine.drug - Abstract
RNA base editing is gaining momentum as an approach to repair mutations, but its application to neurological disease has not been established. We have succeeded in directed transcript editing of a pathological mutation in a mouse model of the neurodevelopmental disease, Rett syndrome. Specifically, we directed editing of a guanosine to adenosine mutation in RNA encoding Methyl CpG Binding Protein 2 (MECP2). Repair was mediated by injecting the hippocampus of juvenile Rett mice with an adeno-associated virus expressing both an engineered enzyme containing the catalytic domain of Adenosine Deaminase Acting on RNA 2 and a Mecp2 targeting guide. After one month, 50% of Mecp2 RNA was recoded in three different hippocampal neuronal subtypes, and the ability of MeCP2 protein to associate with heterochromatin was similarly restored to 50% of wild-type levels. This study represents the first in vivo programmable RNA editing applied to a model of neurological disease.
- Published
- 2020
- Full Text
- View/download PDF
14. Neuronal activity induces glutathione metabolism gene expression in astrocytes
- Author
-
Gail Mandel and James C. McGann
- Subjects
0301 basic medicine ,NF-E2-Related Factor 2 ,Biology ,Receptors, Metabotropic Glutamate ,Hippocampus ,Article ,Flow cytometry ,Transcriptome ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Gene expression ,medicine ,Animals ,Premovement neuronal activity ,Cells, Cultured ,Neurons ,medicine.diagnostic_test ,Glutathione ,Coculture Techniques ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Neurology ,chemistry ,Metabotropic glutamate receptor ,Astrocytes ,Neuron ,Astrocyte - Abstract
The idea that astrocytes provide support for neurons has a long history, but whether neurons play an instructive role in these processes is poorly understood. To address this question, we co-culture astrocytes with genetically labeled neurons, permitting their separation by flow cytometry, and test whether the presence of neurons influences the astrocyte transcriptome. We find that numerous pathways are regulated in the co-cultured astrocytes, in a time-dependent matter coincident with synaptic maturation. In particular, the induction of glutathione metabolic genes is prominent, resulting in increased glutathione production. We show that the induction of the glutathione pathway is mediated by astrocytic metabotropic glutamate receptors. Using a candidate approach, we identify direct binding of the nuclear factor E2-related factor, NRF2, to several of the induced genes. Blocking nuclear accumulation of astrocytic NRF2 abolishes neuron-induced glutathione gene induction and glutathione production. Our results suggest that astrocyte transcriptional and metabolic profiles are tightly coupled to the activity of neurons, consistent with the model that astrocytes dynamically support healthy brain function.
- Published
- 2018
15. Acute and crucial requirement for MeCP2 function upon transition from early to late adult stages of brain maturation
- Author
-
Gail Mandel, Ariel Karten, Christy A. Felice, Minh Vu Chuong Nguyen, Fang Du, and Nurit Ballas
- Subjects
Male ,0301 basic medicine ,Aging ,congenital, hereditary, and neonatal diseases and abnormalities ,Methyl-CpG-Binding Protein 2 ,Physiology ,Mice, Transgenic ,Rett syndrome ,Disease ,Biology ,MECP2 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Genes, X-Linked ,Rett Syndrome ,Genetics ,medicine ,Animals ,Humans ,Juvenile ,Adult stage ,Molecular Biology ,Genetics (clinical) ,Neurons ,Brain ,Articles ,General Medicine ,Anatomy ,medicine.disease ,Phenotype ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Autism spectrum disorder ,Mutation ,Female ,030217 neurology & neurosurgery - Abstract
Germline mutations in the X-linked gene, methyl-CpG-binding protein 2 (MECP2), underlie most cases of Rett syndrome (RTT), an autism spectrum disorder affecting approximately one in 10 000 female live births. The disease is characterized in affected girls by a latent appearance of symptoms between 12 and 18 months of age while boys usually die before the age of two. The nature of the latency is not known, but RTT-like phenotypes are recapitulated in mouse models, even when MeCP2 is removed at different postnatal stages, including juvenile and adolescent stages. Unexpectedly, here, we show that within a very brief developmental window, between 10 (adolescent) and 15 (adult) weeks after birth, symptom initiation and progression upon removal of MeCP2 in male mice transitions from 3 to 4 months to only several days, followed by lethality. We further show that this accelerated development of RTT phenotype and lethality occur at the transition to adult stage (15 weeks of age) and persists thereafter. Importantly, within this abbreviated time frame of days, the brain acquires dramatic anatomical, cellular and molecular abnormalities, typical of classical RTT. This study reveals a new postnatal developmental stage, which coincides with full-brain maturation, where the structure/function of the brain is extremely sensitive to levels of MeCP2 and loss of MeCP2 leads to precipitous collapse of the neuronal networks and incompatibility with life within days.
- Published
- 2016
16. The accessible chromatin landscape of the hippocampus at single-cell resolution
- Author
-
Cole Trapnell, John R. Sinnamon, Sarah A. Vitak, Michael W. Linhoff, Frank J. Steemers, Hannah A. Pliner, Gail Mandel, Andrew Adey, and Kristof A. Torkenczy
- Subjects
0303 health sciences ,Cell type ,Cell ,Promoter ,Computational biology ,Biology ,Hippocampal formation ,Chromatin ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,medicine ,Epigenetics ,030217 neurology & neurosurgery ,DNA ,Transposase ,030304 developmental biology - Abstract
Here we present a comprehensive map of the accessible chromatin landscape of the mouse hippocampus at single-cell resolution. Substantial advances of this work include the optimization of single-cell combinatorial indexing assay for transposase accessible chromatin (sci-ATAC-seq), a software suite,scitools, for the rapid processing and visualization of single-cell combinatorial indexing datasets, and a valuable resource of hippocampal regulatory networks at single-cell resolution. We utilized sci-ATAC-seq to produce 2,346 high-quality single-cell chromatin accessibility maps with a mean unique read count per cell of 29,201 from both fresh and frozen hippocampi, observing little difference in accessibility patterns between the preparations. Using this dataset, we identified eight distinct major clusters of cells representing both neuronal and non-neuronal cell types and characterized the driving regulatory factors and differentially accessible loci that define each cluster. We then applied a recently described co-accessibility framework,Cicero, which identified 146,818 links between promoters and putative distal regulatory DNA. Identified co-accessibility networks showed cell-type specificity, shedding light on key dynamic loci that reconfigure to specify hippocampal cell lineages. Lastly, we carried out an additional sci-ATAC-seq preparation from cultured hippocampal neurons (899 high-quality cells, 43,532 mean unique reads) that revealed substantial alterations in their epigenetic landscape compared to nuclei from hippocampal tissue. This dataset and accompanying analysis tools provide a new resource that can guide subsequent studies of the hippocampus.
- Published
- 2018
17. The accessible chromatin landscape of the murine hippocampus at single-cell resolution
- Author
-
Andrew Adey, Michael W. Linhoff, Frank J. Steemers, Cole Trapnell, Hannah A. Pliner, Ryan M. Mulqueen, John R. Sinnamon, Gail Mandel, Sarah A. Vitak, and Kristof A. Torkenczy
- Subjects
Epigenomics ,Resource ,Sequence analysis ,Cell ,Transposases ,Computational biology ,Hippocampal formation ,Biology ,Hippocampus ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Cell Lineage ,Epigenetics ,Genetics (clinical) ,Transposase ,Cells, Cultured ,030304 developmental biology ,Cell Nucleus ,0303 health sciences ,Neuronal Plasticity ,Pyramidal Cells ,Promoter ,Sequence Analysis, DNA ,Chromatin ,medicine.anatomical_structure ,chemistry ,Single-Cell Analysis ,030217 neurology & neurosurgery ,DNA - Abstract
Here we present a comprehensive map of the accessible chromatin landscape of the mouse hippocampus at single-cell resolution. Substantial advances of this work include the optimization of a single-cell combinatorial indexing assay for transposase accessible chromatin (sci-ATAC-seq); a software suite, scitools, for the rapid processing and visualization of single-cell combinatorial indexing data sets; and a valuable resource of hippocampal regulatory networks at single-cell resolution. We used sci-ATAC-seq to produce 2346 high-quality single-cell chromatin accessibility maps with a mean unique read count per cell of 29,201 from both fresh and frozen hippocampi, observing little difference in accessibility patterns between the preparations. By using this data set, we identified eight distinct major clusters of cells representing both neuronal and nonneuronal cell types and characterized the driving regulatory factors and differentially accessible loci that define each cluster. Within pyramidal neurons, we identified four major clusters, including CA1 and CA3 neurons, and three additional subclusters. We then applied a recently described coaccessibility framework, Cicero, which identified 146,818 links between promoters and putative distal regulatory DNA. Identified coaccessibility networks showed cell-type specificity, shedding light on key dynamic loci that reconfigure to specify hippocampal cell lineages. Lastly, we performed an additional sci-ATAC-seq preparation from cultured hippocampal neurons (899 high-quality cells, 43,532 mean unique reads) that revealed substantial alterations in their epigenetic landscape compared with nuclei from hippocampal tissue. This data set and accompanying analysis tools provide a new resource that can guide subsequent studies of the hippocampus.
- Published
- 2018
18. An RNA Binding Protein Promotes Axonal Integrity in Peripheral Neurons by Destabilizing REST
- Author
-
Francesca Cargnin, Karin Mullendorff, Tamilla Nechiporuk, Deborah J. Stumpo, Perry J. Blackshear, Nurit Ballas, and Gail Mandel
- Subjects
Male ,Nervous system ,RE1-silencing transcription factor ,PC12 Cells ,Mice ,Tristetraprolin ,Ganglia, Spinal ,Gene expression ,medicine ,Animals ,Post-transcriptional regulation ,Cells, Cultured ,Rest (music) ,Mice, Knockout ,Messenger RNA ,Gene knockdown ,biology ,General Neuroscience ,RNA-Binding Proteins ,Articles ,Axons ,Rats ,Cell biology ,Repressor Proteins ,medicine.anatomical_structure ,Peripheral nervous system ,biology.protein ,Female ,Neuroscience - Abstract
The RE1 Silencing Transcription Factor (REST) acts as a governor of the mature neuronal phenotype by repressing a large consortium of neuronal genes in non-neuronal cells. In the developing nervous system, REST is present in progenitors and downregulated at terminal differentiation to promote acquisition of mature neuronal phenotypes. Paradoxically, REST is still detected in some regions of the adult nervous system, but how REST levels are regulated, and whether REST can still repress neuronal genes, is not known. Here, we report that homeostatic levels of REST are maintained in mature peripheral neurons by a constitutive post-transcriptional mechanism. Specifically, using a three-hybrid genetic screen, we identify the RNA binding protein, ZFP36L2, associated previously only with female fertility and hematopoiesis, and show that it regulates REST mRNA stability. Dorsal root ganglia inZfp36l2knock-out mice, or wild-type ganglia expressing ZFP36L2 shRNA, show higher steady-state levels ofRestmRNA and protein, and extend thin and disintegrating axons. This phenotype is due, at least in part, to abnormally elevated REST levels in the ganglia because the axonal phenotype is attenuated by acute knockdown of REST inZfp36l2KO DRG explants. The higher REST levels result in lower levels of target genes, indicating that REST can still fine-tune gene expression through repression. Thus, REST levels are titrated in mature peripheral neurons, in part through a ZFP36L2-mediated post-transcriptional mechanism, with consequences for axonal integrity.
- Published
- 2014
19. Author response: Astrocytic modulation of excitatory synaptic signaling in a mouse model of Rett syndrome
- Author
-
Benjamin Rakela, Paul Brehm, and Gail Mandel
- Published
- 2017
20. Site-directed RNA repair of endogenous Mecp2 RNA in neurons
- Author
-
John R. Sinnamon, Susan Y. Kim, Hiroyuki Nakai, Gail Mandel, Glen M. Corson, John P. Adelman, and Zhen Song
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Adenosine Deaminase ,Methyl-CpG-Binding Protein 2 ,RNA-dependent RNA polymerase ,RNA-binding protein ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Rett Syndrome ,Animals ,Humans ,RNA, Messenger ,Gene ,Transcription factor ,Cells, Cultured ,Neurons ,Messenger RNA ,Multidisciplinary ,RNA ,RNA-Binding Proteins ,RNA repair ,DNA Methylation ,Molecular biology ,nervous system diseases ,Disease Models, Animal ,030104 developmental biology ,PNAS Plus ,RNA editing ,Mutation ,030217 neurology & neurosurgery - Abstract
Rett syndrome (RTT) is a debilitating neurological disorder caused by mutations in the gene encoding the transcription factor Methyl CpG Binding Protein 2 (MECP2). A distinct disorder results from MECP2 gene duplication, suggesting that therapeutic approaches must restore close to normal levels of MECP2. Here, we apply the approach of site-directed RNA editing to repair, at the mRNA level, a disease-causing guanosine to adenosine (G > A) mutation in the mouse MeCP2 DNA binding domain. To mediate repair, we exploit the catalytic domain of Adenosine Deaminase Acting on RNA (ADAR2) that deaminates A to inosine (I) residues that are subsequently translated as G. We fuse the ADAR2 domain, tagged with a nuclear localization signal, to an RNA binding peptide from bacteriophage lambda. In cultured neurons from mice that harbor an RTT patient G > A mutation and express engineered ADAR2, along with an appropriate RNA guide to target the enzyme, 72% of Mecp2 mRNA is repaired. Levels of MeCP2 protein are also increased significantly. Importantly, as in wild-type neurons, the repaired MeCP2 protein is enriched in heterochromatic foci, reflecting restoration of normal MeCP2 binding to methylated DNA. This successful use of site-directed RNA editing to repair an endogenous mRNA and restore protein function opens the door to future in vivo applications to treat RTT and other diseases.
- Published
- 2017
21. REST corepressors RCOR1 and RCOR2 and the repressor INSM1 regulate the proliferation–differentiation balance in the developing brain
- Author
-
Sophia Jeng, Caitlin E. Monaghan, Gail Mandel, Shannon K. McWeeney, Jianxun Wang, Tamilla Nechiporuk, and Michael G. Rosenfeld
- Subjects
0301 basic medicine ,Down-Regulation ,Repressor ,Nerve Tissue Proteins ,RE1-silencing transcription factor ,Mice ,03 medical and health sciences ,Animals ,Transcription factor ,Gene knockout ,Cell Proliferation ,Neurons ,Multidisciplinary ,biology ,Neurogenesis ,Brain ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Phenotype ,Embryonic stem cell ,Up-Regulation ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Repressor Proteins ,RCOR1 ,030104 developmental biology ,PNAS Plus ,biology.protein ,Co-Repressor Proteins ,Neuroscience ,Transcription Factors - Abstract
The transcriptional events that lead to the cessation of neural proliferation, and therefore enable the production of proper numbers of differentiated neurons and glia, are still largely uncharacterized. Here, we report that the transcription factor Insulinoma-associated 1 (INSM1) forms complexes with RE1 Silencing Transcription factor (REST) corepressors RCOR1 and RCOR2 in progenitors in embryonic mouse brain. Mice lacking both RCOR1 and RCOR2 in developing brain die perinatally and generate an abnormally high number of neural progenitors at the expense of differentiated neurons and oligodendrocyte precursor cells. In addition, Rcor1/2 deletion detrimentally affects complex morphological processes such as closure of the interganglionic sulcus. We find that INSM1, a transcription factor that induces cell-cycle arrest, is coexpressed with RCOR1/2 in a subset of neural progenitors and forms complexes with RCOR1/2 in embryonic brain. Further, the Insm1−/− mouse phenocopies predominant brain phenotypes of the Rcor1/2 knockout. A large number of genes are concordantly misregulated in both knockout genotypes, and a majority of the down-regulated genes are targets of REST. Rest transcripts are up-regulated in both knockouts, and reducing transcripts to control levels in the Rcor1/2 knockout partially rescues the defect in interganglionic sulcus closure. Our findings indicate that an INSM1/RCOR1/2 complex controls the balance of proliferation and differentiation during brain development.
- Published
- 2017
22. Corepressor Rcor1 is essential for murine erythropoiesis
- Author
-
Jeffrey W. Tyner, Guang Fan, Shannon K. McWeeney, Gail Mandel, Stuart H. Orkin, William H. Fleming, Marc A. Kerenyi, Huilan Yao, Devorah C. Goldman, Tamilla Nechiporuk, and Sunita Kawane
- Subjects
Myeloid ,Erythroblasts ,Immunology ,Biology ,Biochemistry ,Cytokine Receptor Common beta Subunit ,Mice ,Red Cells, Iron, and Erythropoiesis ,hemic and lymphatic diseases ,medicine ,Animals ,Erythropoiesis ,Myeloid Cells ,Progenitor cell ,Cells, Cultured ,Erythroid Precursor Cells ,Mice, Knockout ,Gene Expression Regulation, Developmental ,Cell Biology ,Hematology ,Embryo, Mammalian ,Receptors, Interleukin-3 ,RCOR1 ,Haematopoiesis ,medicine.anatomical_structure ,Cancer research ,Stem cell ,Co-Repressor Proteins ,Corepressor ,Gene Deletion ,Signal Transduction - Abstract
The corepressor Rcor1 has been linked biochemically to hematopoiesis, but its function in vivo remains unknown. We show that mice deleted for Rcor1 are profoundly anemic and die in late gestation. Definitive erythroid cells from mutant mice arrest at the transition from proerythroblast to basophilic erythroblast. Remarkably, Rcor1 null erythroid progenitors cultured in vitro form myeloid colonies instead of erythroid colonies. The mutant proerythroblasts also aberrantly express genes of the myeloid lineage as well as genes typical of hematopoietic stem cells (HSCs) and/or progenitor cells. The colony-stimulating factor 2 receptor β subunit (Csf2rb), which codes for a receptor implicated in myeloid cytokine signaling, is a direct target for both Rcor1 and the transcription repressor Gfi1b in erythroid cells. In the absence of Rcor1, the Csf2rb gene is highly induced, and Rcor1(-/-) progenitors exhibit CSF2-dependent phospho-Stat5 hypersensitivity. Blocking this pathway can partially reduce myeloid colony formation by Rcor1-deficient erythroid progenitors. Thus, Rcor1 promotes erythropoiesis by repressing HSC and/or progenitor genes, as well as the genes and signaling pathways that lead to myeloid cell fate.
- Published
- 2014
23. Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A
- Author
-
Yu Jung Hsu, Jennifer J. Trowbridge, Jian Xu, Stuart H. Orkin, Serena Hou, Daniel E. Bauer, Marc A. Kerenyi, Thuy D. Vo, Gail Mandel, and Huilan Yao
- Subjects
Proteomics ,Chromatin Immunoprecipitation ,Co-Repressor Proteins ,beta-Globins ,Real-Time Polymerase Chain Reaction ,Mice ,Tandem Mass Spectrometry ,RNA interference ,Cell Line, Tumor ,hemic and lymphatic diseases ,Animals ,Humans ,Gene silencing ,Transcription factor ,Fetal Hemoglobin ,Erythroid Precursor Cells ,Regulation of gene expression ,Multidisciplinary ,biology ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Biological Sciences ,Chromatin ,Repressor Proteins ,Multiprotein Complexes ,biology.protein ,Cancer research ,Demethylase ,RNA Interference ,Carrier Proteins ,Corepressor ,Chromatography, Liquid - Abstract
Reactivation of fetal hemoglobin (HbF) in adults ameliorates the severity of the common β-globin disorders. The transcription factor BCL11A is a critical modulator of hemoglobin switching and HbF silencing, yet the molecular mechanism through which BCL11A coordinates the developmental switch is incompletely understood. Particularly, the identities of BCL11A cooperating protein complexes and their roles in HbF expression and erythroid development remain largely unknown. Here we determine the interacting partner proteins of BCL11A in erythroid cells by a proteomic screen. BCL11A is found within multiprotein complexes consisting of erythroid transcription factors, transcriptional corepressors, and chromatin-modifying enzymes. We show that the lysine-specific demethylase 1 and repressor element-1 silencing transcription factor corepressor 1 (LSD1/CoREST) histone demethylase complex interacts with BCL11A and is required for full developmental silencing of mouse embryonic β-like globin genes and human γ-globin genes in adult erythroid cells in vivo. In addition, LSD1 is essential for normal erythroid development. Furthermore, the DNA methyltransferase 1 (DNMT1) is identified as a BCL11A-associated protein in the proteomic screen. DNMT1 is required to maintain HbF silencing in primary human adult erythroid cells. DNMT1 haploinsufficiency combined with BCL11A deficiency further enhances γ-globin expression in adult animals. Our findings provide important insights into the mechanistic roles of BCL11A in HbF silencing and clues for therapeutic targeting of BCL11A in β-hemoglobinopathies.
- Published
- 2013
24. The REST remodeling complex protects genomic integrity during embryonic neurogenesis
- Author
-
Thomas Floss, Karin Mullendorff, Wolfgang Wurst, Jenny Hsieh, James C. McGann, Tamilla Nechiporuk, and Gail Mandel
- Subjects
0301 basic medicine ,REST complex ,QH301-705.5 ,Science ,Neurogenesis ,Cellular differentiation ,knockout animal ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,Mice ,03 medical and health sciences ,embryology [Brain] ,physiology [Stem Cells] ,Animals ,Biology (General) ,Chromosome separation ,Transcription factor ,transcription factor ,metabolism [Repressor Proteins] ,Rest Complex ,Developmental Biology ,Genomic Instability ,Knockout Animals ,Mouse ,Repression ,Stem Cells ,Transcription Factors ,General Immunology and Microbiology ,General Neuroscience ,Cell Cycle ,Gene targeting ,Cell Differentiation ,General Medicine ,Cell cycle ,genomic instability ,physiology [Neurons] ,RE1-silencing transcription factor ,Molecular biology ,Chromatin ,Cell biology ,Repressor Proteins ,neurogenesis ,030104 developmental biology ,Gene Knockdown Techniques ,Medicine ,repression ,ddc:600 - Abstract
The timely transition from neural progenitor to post-mitotic neuron requires down-regulation and loss of the neuronal transcriptional repressor, REST. Here, we have used mice containing a gene trap in the Rest gene, eliminating transcription from all coding exons, to remove REST prematurely from neural progenitors. We find that catastrophic DNA damage occurs during S-phase of the cell cycle, with long-term consequences including abnormal chromosome separation, apoptosis, and smaller brains. Persistent effects are evident by latent appearance of proneural glioblastoma in adult mice deleted additionally for the tumor suppressor p53 protein (p53). A previous line of mice deleted for REST in progenitors by conventional gene targeting does not exhibit these phenotypes, likely due to a remaining C-terminal peptide that still binds chromatin and recruits co-repressors. Our results suggest that REST-mediated chromatin remodeling is required in neural progenitors for proper S-phase dynamics, as part of its well-established role in repressing neuronal genes until terminal differentiation.
- Published
- 2016
25. Zebrafish model for congenital myasthenic syndrome reveals mechanisms causal to developmental recovery
- Author
-
Jason M. Urban, Nathan R. Nelson, Michael Walogorsky, Rebecca Mongeon, Hua Wen, Gail Mandel, Fumihito Ono, and Paul Brehm
- Subjects
Myasthenic Syndromes, Congenital ,Gene isoform ,medicine.medical_specialty ,Mutation ,Patch-Clamp Techniques ,Multidisciplinary ,Base Sequence ,Biological Sciences ,Congenital myasthenic syndrome ,Biology ,medicine.disease_cause ,medicine.disease ,biology.organism_classification ,Embryonic stem cell ,Disease Models, Animal ,Endocrinology ,Internal medicine ,medicine ,Animals ,Patch clamp ,Receptor ,Zebrafish ,DNA Primers ,Acetylcholine receptor - Abstract
Mutations in muscle ACh receptors cause slow-channel syndrome (SCS) and Escobar syndrome, two forms of congenital myasthenia. SCS is a dominant disorder with mutations reported for all receptor subunits except γ. Escobar syndrome is distinct, with mutations located exclusively in γ, and characterized by developmental improvement of muscle function. The zebrafish mutant line, twister , models SCS in terms of a dominant mutation in the α subunit (α twi ) but shows the behavioral improvement associated with Escobar syndrome. Here, we present a unique electrophysiological study into developmental improvement for a myasthenic syndrome. The embryonic α twi βδγ receptor isoform produces slowly decaying synaptic currents typical of SCS that transit to a much faster decay upon the appearance of adult ε, despite the α twi mutation. Thus, the continued expression of α twi into adulthood is tolerated because of the ε expression and associated recovery, raising the likelihood of unappreciated myasthenic cases that benefit from the γ−ε switch.
- Published
- 2012
26. Acetylcholine Receptor Gating in a Zebrafish Model for Slow-Channel Syndrome
- Author
-
Hua Wen, Paul Brehm, Rebecca Mongeon, Michael Walogorsky, and Gail Mandel
- Subjects
medicine.medical_specialty ,Patch-Clamp Techniques ,Movement ,Xenopus ,Action Potentials ,Gating ,Cholinergic Agonists ,Neurotransmission ,Biology ,Synaptic Transmission ,Cholinergic Antagonists ,Article ,Calcium in biology ,Isomerism ,Internal medicine ,medicine ,Animals ,Receptors, Cholinergic ,Patch clamp ,Muscle, Skeletal ,Receptor ,Zebrafish ,Acetylcholine receptor ,Myasthenic Syndromes, Congenital ,Dose-Response Relationship, Drug ,General Neuroscience ,Quinidine ,Cell biology ,Endocrinology ,Oocytes ,Cholinergic ,Calcium ,Channelopathies ,Ion Channel Gating ,Acetylcholine ,medicine.drug - Abstract
Slow channel syndrome (SCS) is an autosomal dominant disease resulting from mutations in muscle acetylcholine receptor (AChR) subunits. The associated fatigue and muscle degeneration are proposed to result from prolonged synaptic responses that overload intracellular calcium. Single channel studies on reconstituted receptors bearing human mutations indicate that the prolonged responses result from an increase in receptor open duration and, in some cases, increased sensitivity to acetylcholine (ACh). We show that both of these aberrant receptor properties are recapitulated in heterozygotic zebrafish bearing an L258P mutation in the α subunit, thus affording the unique opportunity to compare the single channel properties of mutant receptors to the synaptic currents in vivo. Whole cell recordings revealed synaptic currents that decayed along a multi-exponential time course, reflecting receptors containing mixtures of wild type and mutant α subunits. Treatment with quinidine, an open channel blocker used to treat the human disorder, restored fast synaptic current kinetics and the ability to swim. Quinidine block also revealed that mutant receptors generate a large steady state current in the absence of ACh. The spontaneous openings reflected a destabilization of the closed state, leading to an apparent increase in the sensitivity of these receptors to ACh. The effective block by quinidine on synaptic currents as well as non-liganded openings points to dual sources for the calcium-dependent myopathy in certain forms of SCS.
- Published
- 2012
27. Nonequivalent release sites govern synaptic depression
- Author
-
Hua Wen, Gail Mandel, Paul Brehm, and Matthew J. McGinley
- Subjects
0301 basic medicine ,Time Factors ,Green Fluorescent Proteins ,Neuromuscular transmission ,Neuromuscular Junction ,Stimulation ,Mice, Transgenic ,Biology ,Synaptic vesicle ,Neuromuscular junction ,Synapse ,03 medical and health sciences ,0302 clinical medicine ,Neuroplasticity ,medicine ,Animals ,Patch clamp ,Zebrafish ,Probability ,Motor Neurons ,Multidisciplinary ,Neuronal Plasticity ,Reproducibility of Results ,Electric Stimulation ,030104 developmental biology ,medicine.anatomical_structure ,PNAS Plus ,Synaptic plasticity ,Synapses ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Synaptic depression is prominent among synapses, but the underlying mechanisms remain uncertain. Here, we use paired patch clamp recording to study neuromuscular transmission between the caudal primary motor neuron and target skeletal muscle in zebrafish. This synapse has an unusually low number of release sites, all with high probabilities of release in response to low-frequency stimulation. During high-frequency stimulation, the synapse undergoes short-term depression and reaches steady-state levels of transmission that sustain the swimming behavior. To determine the release parameters underlying this steady state, we applied variance analysis. Our analysis revealed two functionally distinct subclasses of release sites differing by over 60-fold in rates of vesicle reloading. A slow reloading class requires seconds to recover and contributes to depression onset but not the steady-state transmission. By contrast, a fast reloading class recovers within tens of milliseconds and is solely responsible for steady-state transmission. Thus, in contrast to most current models that assign levels of steady-state depression to vesicle availability, our findings instead assign this function to nonuniform release site kinetics. The duality of active-site properties accounts for the highly nonlinear dependence of steady-state depression levels on frequency.
- Published
- 2015
28. Repressor element 1 silencing transcription factor (REST) controls radial migration and temporal neuronal specification during neocortical development
- Author
-
Christopher G. Fiondella, Gail Mandel, Diane D. Lu, Joseph J. LoTurco, Nurit Ballas, and Matthew V. Covey
- Subjects
Doublecortin Domain Proteins ,Chromatin Immunoprecipitation ,DNA, Complementary ,Neurogenesis ,Cellular differentiation ,Blotting, Western ,Genetic Vectors ,Repressor ,Neocortex ,Nerve Tissue Proteins ,Biology ,Cell Line ,Mice ,Cell Movement ,Animals ,Humans ,Gene silencing ,Transcription factor ,DNA Primers ,Progenitor ,Mice, Knockout ,Microscopy, Confocal ,Multidisciplinary ,Neuropeptides ,Cell Differentiation ,Biological Sciences ,Immunohistochemistry ,Molecular biology ,Neural stem cell ,Doublecortin ,Cell biology ,Repressor Proteins ,Electroporation ,biology.protein ,Co-Repressor Proteins ,Microtubule-Associated Proteins - Abstract
Neurogenesis requires mechanisms that coordinate early cell-fate decisions, migration, and terminal differentiation. Here, we show that the transcriptional repressor, repressor element 1 silencing transcription factor (REST), regulates radial migration and the timing of neural progenitor differentiation during neocortical development, and that the regulation is contingent upon differential REST levels. Specifically, a sustained presence of REST blocks migration and greatly delays—but does not prevent—neuronal differentiation, resulting in a subcortical band heterotopia-like phenotype, reminiscent of loss of doublecortin. We further show that doublecortin is a direct gene target of REST, and that its overexpression rescues, at least in part, the aberrant phenotype caused by persistent presence of REST. Our studies support the view that the targeted down-regulation of REST to low levels in neural progenitors, and its subsequent disappearance during neurogenesis, is critical for timing the spatiotemporal transition of neural progenitor cells to neurons.
- Published
- 2011
29. An acetylcholine receptor lacking both γ and ε subunits mediates transmission in zebrafish slow muscle synapses
- Author
-
Paul Brehm, Michael Walogorsky, Gail Mandel, Jason M. Urban, Fumihito Ono, and Rebecca Mongeon
- Subjects
Physiology ,Protein subunit ,Molecular Sequence Data ,Xenopus ,Article ,Xenopus laevis ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Receptors, Cholinergic ,Amino Acid Sequence ,Muscle, Skeletal ,Receptor ,Zebrafish ,030304 developmental biology ,Acetylcholine receptor ,0303 health sciences ,biology ,Skeletal muscle ,Zebrafish Proteins ,biology.organism_classification ,Rats ,Cell biology ,Kinetics ,Protein Subunits ,Nicotinic acetylcholine receptor ,medicine.anatomical_structure ,Synapses ,Oocytes ,Cholinergic ,Female ,Sequence Alignment ,Neuroscience ,030217 neurology & neurosurgery ,Acetylcholine ,medicine.drug - Abstract
Fast and slow skeletal muscle types in larval zebrafish can be distinguished by a fivefold difference in the time course of their synaptic decay. Single-channel recordings indicate that this difference is conferred through kinetically distinct nicotinic acetylcholine receptor (AChR) isoforms. The underlying basis for this distinction was explored by cloning zebrafish muscle AChR subunit cDNAs and expressing them in Xenopus laevis oocytes. Measurements of single-channel conductance and mean open burst duration assigned α(2)βδε to fast muscle synaptic current. Contrary to expectations, receptors composed of only αβδ subunits (presumed to be α(2)βδ(2) receptors) recapitulated the kinetics and conductance of slow muscle single-channel currents. Additional evidence in support of γ/ε-less receptors as mediators of slow muscle synapses was reflected in the inward current rectification of heterologously expressed α(2)βδ(2) receptors, a property normally associated with neuronal-type nicotinic receptors. Similar rectification was reflected in both single-channel and synaptic currents in slow muscle, distinguishing them from fast muscle. The final evidence for α(2)βδ(2) receptors in slow muscle was provided by our ability to convert fast muscle synaptic currents to those of slow muscle by knocking down ε subunit expression in vivo. Thus, for the first time, muscle synaptic function can be ascribed to a receptor isoform that is composed of only three different subunits. The unique functional features offered by the α(2)βδ(2) receptor likely play a central role in mediating the persistent contractions characteristic to this muscle type.
- Published
- 2011
30. A role for glia in the progression of Rett’s syndrome
- Author
-
Brian K. Kaspar, Daniel T. Lioy, Gail Mandel, Saurabh K. Garg, Frank Kirchhoff, Jacob Raber, Petra G. Hirrlinger, Kevin D. Foust, Nurit Ballas, Caitlin E. Monaghan, and John M. Bissonnette
- Subjects
Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Methyl-CpG-Binding Protein 2 ,Mutant ,Rett syndrome ,Neuropathology ,Anxiety ,Motor Activity ,Biology ,Article ,MECP2 ,Mice ,Degenerative disease ,mental disorders ,Rett Syndrome ,medicine ,Animals ,Loss function ,Neurons ,Regulation of gene expression ,Multidisciplinary ,Behavior, Animal ,medicine.disease ,nervous system diseases ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Gene Expression Regulation ,Astrocytes ,Vesicular Glutamate Transport Protein 1 ,Disease Progression ,Neuroglia ,Female ,Neuroscience - Abstract
Rett's syndrome (RTT) is an X-chromosome-linked autism spectrum disorder caused by loss of function of the transcription factor methyl-CpG-binding protein 2 (MeCP2). Although MeCP2 is expressed in most tissues, loss of MeCP2 expression results primarily in neurological symptoms. Earlier studies suggested the idea that RTT is due exclusively to loss of MeCP2 function in neurons. Although defective neurons clearly underlie the aberrant behaviours, we and others showed recently that the loss of MECP2 from glia negatively influences neurons in a non-cell-autonomous fashion. Here we show that in globally MeCP2-deficient mice, re-expression of Mecp2 preferentially in astrocytes significantly improved locomotion and anxiety levels, restored respiratory abnormalities to a normal pattern, and greatly prolonged lifespan compared to globally null mice. Furthermore, restoration of MeCP2 in the mutant astrocytes exerted a non-cell-autonomous positive effect on mutant neurons in vivo, restoring normal dendritic morphology and increasing levels of the excitatory glutamate transporter VGLUT1. Our study shows that glia, like neurons, are integral components of the neuropathology of RTT, and supports the targeting of glia as a strategy for improving the associated symptoms.
- Published
- 2011
31. microRNA-132 regulates dendritic growth and arborization of newborn neurons in the adult hippocampus
- Author
-
Gary L. Westbrook, Xiaolu A. Cambronne, Daniel T. Lioy, Richard H. Goodman, Barbara H. Leighton, Bryan W. Luikart, Gail Mandel, and Stephen T. Magill
- Subjects
Neurogenesis ,Cre recombinase ,Mice, Transgenic ,Biology ,Hippocampal formation ,CREB ,Hippocampus ,Gene Knockout Techniques ,Mice ,miR-132 ,Cell Line, Tumor ,Animals ,Humans ,Transcription factor ,Neurons ,Microscopy, Confocal ,Multidisciplinary ,Dentate gyrus ,Cell Differentiation ,Dendrites ,Biological Sciences ,Flow Cytometry ,CREB-Binding Protein ,Immunohistochemistry ,Molecular biology ,Cell biology ,MicroRNAs ,Gene Expression Regulation ,biology.protein ,Signal transduction ,Signal Transduction - Abstract
Newborn neurons in the dentate gyrus of the adult hippocampus rely upon cAMP response element binding protein (CREB) signaling for their differentiation into mature granule cells and their integration into the dentate network. Among its many targets, the transcription factor CREB activates expression of a gene locus that produces two microRNAs, miR-132 and miR-212. In cultured cortical and hippocampal neurons, miR-132 functions downstream from CREB to mediate activity-dependent dendritic growth and spine formation in response to a variety of signaling pathways. To investigate whether miR-132 and/or miR-212 contribute to the maturation of dendrites in newborn neurons in the adult hippocampus, we inserted LoxP sites surrounding the miR-212/132 locus and specifically targeted its deletion by stereotactically injecting a retrovirus expressing Cre recombinase. Deletion of the miR-212/132 locus caused a dramatic decrease in dendrite length, arborization, and spine density. The miR-212/132 locus may express up to four distinct microRNAs, miR-132 and -212 and their reverse strands miR-132* and -212*. Using ratiometric microRNA sensors, we determined that miR-132 is the predominantly active product in hippocampal neurons. We conclude that miR-132 is required for normal dendrite maturation in newborn neurons in the adult hippocampus and suggest that this microRNA also may participate in other examples of CREB-mediated signaling.
- Published
- 2010
32. A New Binding Motif for the Transcriptional Repressor REST Uncovers Large Gene Networks Devoted to Neuronal Functions
- Author
-
Stefanie J. Otto, John Hover, Richard H. Goodman, Soren Impey, Gail Mandel, Sean R. McCorkle, John J. Dunn, Jong Jin Han, Cecilia Conaco, and Gregory S. Yochum
- Subjects
Neurons ,Genetics ,Binding Sites ,biology ,Cell adhesion molecule ,Gene Expression Profiling ,General Neuroscience ,Amino Acid Motifs ,Repressor ,Articles ,RE1-silencing transcription factor ,Cell Line ,Cell biology ,Chromatin ,Repressor Proteins ,Gene expression profiling ,Mice ,biology.protein ,Animals ,Gene silencing ,Gene Regulatory Networks ,Gene ,Transcription factor ,Transcription Factors - Abstract
The repressor element 1 (RE1) silencing transcription factor (REST) helps preserve the identity of nervous tissue by silencing neuronal genes in non-neural tissues. Moreover, in an epithelial model of tumorigenesis, loss of REST function is associated with loss of adhesion, suggesting the aberrant expression of REST-controlled genes encoding this property. To date, no adhesion molecules under REST control have been identified. Here, we used serial analysis of chromatin occupancy to perform genome-wide identification of REST-occupied target sequences (RE1 sites) in a kidney cell line. We discovered novel REST-binding motifs and found that the number of RE1 sites far exceeded previous estimates. A large family of targets encoding adhesion proteins was identified, as were genes encoding signature proteins of neuroendocrine tumors. Unexpectedly, genes considered exclusively non-neuronal also contained an RE1 motif and were expressed in neurons. This supports the model that REST binding is a critical determinant of neuronal phenotype.
- Published
- 2007
33. Author response: The REST remodeling complex protects genomic integrity during embryonic neurogenesis
- Author
-
Wolfgang Wurst, Gail Mandel, Jenny Hsieh, Thomas Floss, Karin Mullendorff, Tamilla Nechiporuk, and James C. McGann
- Subjects
Neurogenesis ,Biology ,Neuroscience ,Embryonic stem cell ,Rest (music) - Published
- 2015
34. The Corepressor Rcor1 Is Essential for Normal Myeloerythroid Lineage Differentiation
- Author
-
William H. Fleming, Devorah C. Goldman, Gail Mandel, Guang Fan, and Huilan Yao
- Subjects
Neutrophils ,Mice, Transgenic ,Biology ,Monocytes ,Article ,Mice ,Megakaryocyte ,Erythroid Cells ,medicine ,Animals ,Cell Lineage ,Progenitor cell ,Erythroid Precursor Cells ,Mice, Knockout ,Monocyte ,GATA2 ,Cell Differentiation ,Cell Biology ,medicine.disease ,RCOR1 ,Mice, Inbred C57BL ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Cancer research ,Molecular Medicine ,Bone marrow ,Co-Repressor Proteins ,Developmental Biology - Abstract
Based on its physical interactions with histone-modifying enzymes, the transcriptional corepressor Rcor1 has been implicated in the epigenetic regulation blood cell development. Previously, we have demonstrated that Rcor1 is essential for the maturation of definitive erythroid cells and fetal survival. To determine the functional role of Rcor1 in steady-state hematopoiesis in the adult, we used a conditional knockout approach. Here, we show that the loss of Rcor1 expression results in the rapid onset of severe anemia due to a complete, cell autonomous block in the maturation of committed erythroid progenitors. By contrast, both the frequency of megakaryocyte progenitors and their capacity to produce platelets were normal. Although the frequency of common lymphoid progenitors and T cells was not altered, B cells were significantly reduced and showed increased apoptosis. However, Rcor1-deficient bone marrow sustained normal levels of B-cells following transplantation, indicating a non-cell autonomous requirement for Rcor1 in B-cell survival. Evaluation of the myelomonocytic lineage revealed an absence of mature neutrophils and a significant increase in the absolute number of monocytic cells. Rcor1-deficient monocytes were less apoptotic and showed ∼100-fold more colony-forming activity than their normal counterparts, but did not give rise to leukemia. Moreover, Rcor1−/− monocytes exhibited extensive, cytokine-dependent self-renewal and overexpressed genes associated with hematopoietic stem/progenitor cell expansion including Gata2, Meis1, and Hoxa9. Taken together, these data demonstrate that Rcor1 is essential for the normal differentiation of myeloerythroid progenitors and for appropriately regulating self-renewal activity in the monocyte lineage. Stem Cells 2015;33:3304–3314
- Published
- 2015
35. Epigenetic Mechanisms and Gene Networks in the Nervous System
- Author
-
William Renthal, Arvind Kumar, Gail Mandel, Soren Impey, Christine M. Colvis, Eric J. Nestler, Mark Mayford, Richard H. Goodman, Jonathan D. Pollock, Frances A. Champagne, John J. Dunn, Edward Korzus, and David E. H. Theobald
- Subjects
Models, Molecular ,Nervous system ,Substance-Related Disorders ,Gene regulatory network ,Computational biology ,Environment ,Biology ,Nervous System ,complex mixtures ,Genome ,Chromatin remodeling ,Epigenesis, Genetic ,Histones ,medicine ,Animals ,Gene silencing ,Gene Silencing ,Epigenetics ,Transcription factor ,Neurons ,Genetics ,Symposia and Mini-Symposia ,General Neuroscience ,DNA Methylation ,Chromatin Assembly and Disassembly ,equipment and supplies ,medicine.anatomical_structure ,DNA methylation ,bacteria ,Transcription Factors - Abstract
Adaptation to the environment is one of the fundamental regulatory processes in biology and is found among both simple and complex organisms. In a changing environment, simple organisms enhance species survival by high rates of spontaneous mutation achieved by several means: short maturation rates
- Published
- 2005
36. The many faces of REST oversee epigenetic programming of neuronal genes
- Author
-
Gail Mandel and Nurit Ballas
- Subjects
Neurons ,Pluripotent Stem Cells ,Nervous system ,Epigenetic regulation of neurogenesis ,biology ,General Neuroscience ,Cellular differentiation ,Neurogenesis ,Gene Expression Regulation, Developmental ,Cell Differentiation ,RE1-silencing transcription factor ,Epigenesis, Genetic ,Repressor Proteins ,medicine.anatomical_structure ,nervous system ,medicine ,biology.protein ,Animals ,Humans ,Cell Lineage ,Epigenetics ,Induced pluripotent stem cell ,Neural cell ,Neuroscience ,Transcription Factors - Abstract
Nervous system development relies on a complex signaling network to engineer the orderly transitions that lead to the acquisition of a neural cell fate. Progression from the non-neuronal pluripotent stem cell to a restricted neural lineage is characterized by distinct patterns of gene expression, particularly the restriction of neuronal gene expression to neurons. Concurrently, cells outside the nervous system acquire and maintain a non-neuronal fate that permanently excludes expression of neuronal genes. Studies of the transcriptional repressor REST, which regulates a large network of neuronal genes, provide a paradigm for elucidating the link between epigenetic mechanisms and neurogenesis. REST orchestrates a set of epigenetic modifications that are distinct between non-neuronal cells that give rise to neurons and those that are destined to remain as nervous system outsiders.
- Published
- 2005
37. A Genetic Screen for Candidate Tumor Suppressors Identifies REST
- Author
-
Thomas F. Westbrook, Anthony C. Liang, Eric S. Martin, Stephen J. Elledge, Jean J. Zhao, Thomas M. Roberts, Ronald A. DePinho, Gail Mandel, Yumei Leng, Gregory J. Hannon, Michael R. Schlabach, Bin Feng, and Lynda Chin
- Subjects
Candidate gene ,RE1-silencing transcription factor ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Malignant transformation ,Frameshift mutation ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,Cell Line, Tumor ,medicine ,Humans ,PTEN ,Genes, Tumor Suppressor ,Genetic Testing ,Epigenetics ,Cells, Cultured ,Phosphoinositide-3 Kinase Inhibitors ,030304 developmental biology ,Genetics ,0303 health sciences ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Epithelial Cells ,3. Good health ,Repressor Proteins ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,RNA Interference ,Carcinogenesis ,Signal Transduction ,Transcription Factors ,Genetic screen - Abstract
SummaryTumorigenesis is a multistep process characterized by a myriad of genetic and epigenetic alterations. Identifying the causal perturbations that confer malignant transformation is a central goal in cancer biology. Here we report an RNAi-based genetic screen for genes that suppress transformation of human mammary epithelial cells. We identified genes previously implicated in proliferative control and epithelial cell function including two established tumor suppressors, TGFBR2 and PTEN. In addition, we uncovered a previously unrecognized tumor suppressor role for REST/NRSF, a transcriptional repressor of neuronal gene expression. Array-CGH analysis identified REST as a frequent target of deletion in colorectal cancer. Furthermore, we detect a frameshift mutation of the REST gene in colorectal cancer cells that encodes a dominantly acting truncation capable of transforming epithelial cells. Cells lacking REST exhibit increased PI(3)K signaling and are dependent upon this pathway for their transformed phenotype. These results implicate REST as a human tumor suppressor and provide a novel approach to identifying candidate genes that suppress the development of human cancer.
- Published
- 2005
38. Components of the REST/CoREST/histone deacetylase repressor complex are disrupted, modified, and translocated in HSV-1-infected cells
- Author
-
Haidong Gu, Yu Liang, Bernard Roizman, and Gail Mandel
- Subjects
Gene Expression Regulation, Viral ,Viral protein ,Ubiquitin-Protein Ligases ,viruses ,Immunoblotting ,Repressor ,Histone Deacetylase 1 ,Nerve Tissue Proteins ,Herpesvirus 1, Human ,Biology ,Cell Fractionation ,medicine.disease_cause ,Histone Deacetylases ,Immediate early protein ,Cell Line ,Immediate-Early Proteins ,medicine ,Humans ,Immunoprecipitation ,Gene silencing ,Phosphorylation ,Regulation of gene expression ,Microscopy, Confocal ,Multidisciplinary ,Transfection ,Biological Sciences ,Molecular biology ,DNA-Binding Proteins ,Repressor Proteins ,Protein Transport ,Multiprotein Complexes ,Histone deacetylase ,Co-Repressor Proteins ,Corepressor ,Transcription Factors - Abstract
The infected cell protein (ICP)0 enables gene expression and the replication of herpes simplex virus (HSV)-1 in cells infected at low multiplicities and enhances the expression of genes introduced into cells by transfection or infection. We report that a short sequence of ICP0 is similar to a sequence in the amino terminus of CoREST, a corepressor that exists in complexes with the repressor REST and histone deacetylases (HDACs) 1 or 2 to repress cellular gene expression. In wild-type-virus-infected cells, HDAC1 dissociates from the CoREST/REST complex, CoREST and HDAC1 are phosphorylated by a process mediated by viral protein kinases, and CoREST and HDAC1 are partially translocated to the cytoplasm. In cells infected with a virus mutant (ΔICP4), in which ICP0 accumulates, but post-α gene expression is blocked, HDAC1 is dissociated from the CoREST/REST complex, but translocation to the cytoplasm does not occur. After infection with a mutant virus from which ICP0 is deleted, the complex remains intact, but, under conditions of productive infection, the complex is partially translocated to the cytoplasm. These results suggest that, at low multiplicities of infection, ICP0 blocks CoREST-mediated silencing of viral genes by dissociation of HDAC1, whereas subsequent modifications and translocation of the components of the complex are the functions of other viral gene products made later in infection.
- Published
- 2005
39. Defining the CREB Regulon
- Author
-
Jeremy M. Boss, Shannon K. McWeeney, Gail Mandel, Richard H. Goodman, John J. Dunn, Gregory S. Yochum, Jami M. Dwyer, Soren Impey, Hyunjoo Cha-Molstad, and Sean R. McCorkle
- Subjects
Genetics ,DNA binding site ,ATF/CREB ,Transcription (biology) ,Biochemistry, Genetics and Molecular Biology(all) ,biology.protein ,Promoter ,Biology ,CREB ,Chromatin immunoprecipitation ,Transcription factor ,General Biochemistry, Genetics and Molecular Biology ,Chromatin - Abstract
The CREB transcription factor regulates differentiation, survival, and synaptic plasticity. The complement of CREB targets responsible for these responses has not been identified, however. We developed a novel approach to identify CREB targets, termed serial analysis of chromatin occupancy (SACO), by combining chromatin immunoprecipitation (ChIP) with a modification of SAGE. Using a SACO library derived from rat PC12 cells, we identified ∼41,000 genomic signature tags (GSTs) that mapped to unique genomic loci. CREB binding was confirmed for all loci supported by multiple GSTs. Of the 6302 loci identified by multiple GSTs, 40% were within 2 kb of the transcriptional start of an annotated gene, 49% were within 1 kb of a CpG island, and 72% were within 1 kb of a putative cAMP-response element (CRE). A large fraction of the SACO loci delineated bidirectional promoters and novel antisense transcripts. This study represents the most comprehensive definition of transcription factor binding sites in a metazoan species.
- Published
- 2004
- Full Text
- View/download PDF
40. Persistent Electrical Coupling and Locomotory Dysfunction in the Zebrafish Mutantshocked
- Author
-
Paul Brehm, Fumihito Ono, Meng Wang, Michelle R. Gleason, Gail Mandel, Victor M. Luna, and Julia E. Dallman
- Subjects
Patch-Clamp Techniques ,Physiology ,Connexins ,Synapse ,medicine ,Animals ,Myocyte ,Patch clamp ,Evoked Potentials ,Zebrafish ,biology ,Chemistry ,General Neuroscience ,Gap junction ,Depolarization ,Motor neuron ,biology.organism_classification ,Electrophysiology ,Phenotype ,medicine.anatomical_structure ,Connexin 43 ,Mutation ,Nervous System Diseases ,Neuroscience ,Locomotion - Abstract
On initial formation of neuromuscular junctions, slow synaptic signals interact through an electrically coupled network of muscle cells. After the developmental onset of muscle excitability and the transition to fast synaptic responses, electrical coupling diminishes. No studies have revealed the functional importance of the electrical coupling or its precisely timed loss during development. In the mutant zebrafish shocked ( sho) electrical coupling between fast muscle cells persists beyond the time that it would normally disappear in wild-type fish. Recordings from sho indicate that muscle depolarization in response to motor neuron stimulation remains slow due to the low-pass filter characteristics of the coupled network of muscle cells. Our findings suggest that the resultant prolonged muscle depolarizations contribute to the premature termination of swimming in sho and the delayed acquisition of the normally rapid touch-triggered movements. Thus the benefits of gap junctions during early synapse development likely become a liability if not inactivated by the time that muscle would normally achieve fast autonomous function.
- Published
- 2004
41. A Conserved Role But Different Partners for the Transcriptional Corepressor CoREST in Fly and Mammalian Nervous System Formation
- Author
-
Andrew R. Bassett, Andrew Travers, Julia E. Dallman, Gail Mandel, and Janet Allopenna
- Subjects
Central Nervous System ,Development/Plasticity/Repair ,Repressor ,Nerve Tissue Proteins ,Cell Line ,Mice ,Two-Hybrid System Techniques ,Animals ,Drosophila Proteins ,Humans ,Gene silencing ,Amino Acid Sequence ,Psychological repression ,Gene ,Transcription factor ,Conserved Sequence ,Neurons ,Genetics ,biology ,General Neuroscience ,biology.organism_classification ,DNA-Binding Proteins ,Repressor Proteins ,RCOR1 ,Drosophila melanogaster ,Larva ,Drosophila ,Co-Repressor Proteins ,Corepressor ,Transcription Factors - Abstract
Identification of conserved proteins that act to establish the neuronal phenotype has relied predominantly on structural homologies of the underlying genes. In the case of the repressor element 1 silencing transcription factor (REST), a central player in blocking the neuronal phenotype in vertebrate non-neural tissue, the invertebrate homolog is absent, raising the possibility that distinct strategies are used to establish the CNS of invertebrates. Using a yeast two-hybrid screen designed specifically to identify functional analogs of REST, we show thatDrosophila melanogasteruses a strategy that is functionally similar to, but appears to have evolved independently of, REST. The gene at the center of the strategy in flies encodes the repressor Tramtrack88 (Ttk88), a protein with no discernable homology to REST but that nonetheless is able to interact with the same transcriptional partners. Ttk88 uses the REST corepressorDrosophilaCoREST to coordinately regulate a set of genes encoding the same neuronal hallmarks that are regulated by REST in vertebrates. Our findings indicate that repression is an important mechanism for regulating neuronal phenotype across phyla and suggest that co-option of a similar corepressor complex occurred to restrict expression of genes critical for neuronal function to a compartmentalized nervous system.
- Published
- 2004
42. Engrailed-1 Expression Marks a Primitive Class of Inhibitory Spinal Interneuron
- Author
-
Shin-ichi Higashijima, Mark A. Masino, Joseph R. Fetcho, and Gail Mandel
- Subjects
Embryo, Nonmammalian ,Patch-Clamp Techniques ,Time Factors ,Interneuron ,Glycine ,Nerve Tissue Proteins ,Behavioral/Systems/Cognitive ,Biology ,Inhibitory postsynaptic potential ,Glycine Plasma Membrane Transport Proteins ,Interneurons ,Postsynaptic potential ,medicine ,Animals ,Glycine receptor ,In Situ Hybridization ,Zebrafish ,Spinal interneuron ,Microscopy, Confocal ,musculoskeletal, neural, and ocular physiology ,General Neuroscience ,fungi ,Zebrafish Proteins ,Spinal cord ,Amino Acid Transport Systems, Neutral ,medicine.anatomical_structure ,Spinal Cord ,nervous system ,Larva ,GDF7 ,Glycine transporter 2 ,biology.protein ,Neuroscience ,Transcription Factors - Abstract
Studies in chicks and mice have suggested that transcription factors mark functional subtypes of interneurons in the developing spinal cord. We used genetic, morphological, and physiological studies to test this proposed association in zebrafish. We found that Engrailed-1 expression uniquely marks a class of ascending interneurons, called circumferential ascending (CiA) interneurons, with ipsilateral axonal projections in both motor and sensory regions of spinal cord. These cells express the glycine transporter 2 gene and are the only known ipsilateral interneurons positive for this marker of inhibitory transmission. Patch recordings show that the CiA neurons are rhythmically active during swimming. Pairwise recordings from the CiA interneurons and postsynaptic cells reveal that the Engrailed-1 neurons produce monosynaptic, strychnine-sensitive inhibition of dorsal sensory interneurons and also inhibit more ventral neurons, including motoneurons and descending interneurons. We conclude that Engrailed-1 expression marks a class of inhibitory interneuron that seems to provide all of the ipsilateral glycinergic inhibition in the spinal cord of embryonic and larval fish. Individual Engrailed-1-positive cells are multifunctional, playing roles in both sensory gating and motor pattern generation. This primitive cell type may have given rise to several, more specialized glycinergic inhibitory interneurons in birds and mammals. Our data support the view that the subdivision of spinal cord into different regions by transcription factors defines a primitive functional organization of spinal interneurons that formed a developmental and evolutionary foundation on which more complex systems were built.
- Published
- 2004
43. Corepressor-Dependent Silencing of Chromosomal Regions Encoding Neuronal Genes
- Author
-
Pavel A. Pevzner, Sing-Hoi Sze, Robert M. Burgess, Josh Chenoweth, Phillip Schwartz, Victoria V. Lunyak, Gail Mandel, Gratien G. Prefontaine, Michael G. Rosenfeld, Charles A. Nelson, and Christopher K. Glass
- Subjects
Chromosomal Proteins, Non-Histone ,Methyl-CpG-Binding Protein 2 ,Repressor ,Nerve Tissue Proteins ,RE1-silencing transcription factor ,Transfection ,Chromosomes ,Histone Deacetylases ,Sodium Channels ,Cell Line ,Mice ,Animals ,Chromosomes, Human ,Humans ,Gene silencing ,Gene Silencing ,Nerve Growth Factors ,Promoter Regions, Genetic ,Psychological repression ,Transcription factor ,Neurons ,Genetics ,Binding Sites ,NAV1.2 Voltage-Gated Sodium Channel ,Multidisciplinary ,Models, Genetic ,biology ,Gene Expression Profiling ,Calcium-Binding Proteins ,Intracellular Signaling Peptides and Proteins ,Computational Biology ,Membrane Proteins ,DNA Methylation ,Protein Structure, Tertiary ,Rats ,DNA-Binding Proteins ,Repressor Proteins ,RCOR1 ,Gene Expression Regulation ,Microtubule Proteins ,biology.protein ,Stathmin ,CpG Islands ,Histone deacetylase ,Carrier Proteins ,Co-Repressor Proteins ,Corepressor ,Transcription Factors - Abstract
The molecular mechanisms by which central nervous system–specific genes are expressed only in the nervous system and repressed in other tissues remain a central issue in developmental and regulatory biology. Here, we report that the zinc-finger gene-specific repressor element RE-1 silencing transcription factor/neuronal restricted silencing factor (REST/NRSF) can mediate extraneuronal restriction by imposing either active repression via histone deacetylase recruitment or long-term gene silencing using a distinct functional complex. Silencing of neuronal-specific genes requires the recruitment of an associated corepressor, CoREST, that serves as a functional molecular beacon for the recruitment of molecular machinery that imposes silencing across a chromosomal interval, including transcriptional units that do not themselves contain REST/NRSF response elements.
- Published
- 2002
44. REST Repression of Neuronal Genes Requires Components of the hSWI·SNF Complex
- Author
-
Gail Mandel, Josh G. Chenoweth, D W Rose, Michael G. Rosenfeld, Mary E. Anderson, Elena Battaglioli, and María Estela Andrés
- Subjects
Chromosomal Proteins, Non-Histone ,Molecular Sequence Data ,Repressor ,Nerve Tissue Proteins ,Biology ,Biochemistry ,Histone Deacetylases ,Sodium Channels ,Chromatin remodeling ,Cell Line ,Two-Hybrid System Techniques ,Animals ,Humans ,SIN3A ,Amino Acid Sequence ,Promoter Regions, Genetic ,Molecular Biology ,Psychological repression ,Neurons ,Reporter gene ,NAV1.2 Voltage-Gated Sodium Channel ,Sequence Homology, Amino Acid ,Cell Biology ,Molecular biology ,Rats ,Repressor Proteins ,Alternative Splicing ,Histone ,Gene Expression Regulation ,Acetylation ,biology.protein ,Histone deacetylase activity ,Transcription Factors - Abstract
A function of the transcription factor REST is to block the expression of neuronal phenotypic traits in non-neuronal cells. Previous studies have shown that REST-mediated repression requires histone deacetylase activity and that recruitment of deacetylases is mediated by two co-repressors, Sin3A and CoREST. In this study, we show that a repressor domain in CoREST interacts with BRG1-associated factor (BAF) 57, a component of the hSWI.SNF complex. In vivo, BAF57 occupies the neuronal sodium channel gene (Nav1.2) promoter, and targeting to this gene requires REST. In addition to BAF57, the ATPase BRG1 and BAF170, other members of the hSWI.SNF complex, are also present in the REST.CoREST repressor complex. Microinjection of specific antibodies against BRG1, BAF57, or BAF170 into Rat1 fibroblasts relieves repression of RE1 reporter genes. Together, our data suggest that ATP-dependent chromatin remodeling, as well as histone deacetylation, is needed for REST-mediated repression.
- Published
- 2002
45. The Zebrafish Motility Mutanttwitch onceReveals New Roles for Rapsyn in Synaptic Function
- Author
-
Shin-ichi Higashijima, Anatoly Shcherbatko, Fumihito Ono, Gail Mandel, and Paul Brehm
- Subjects
Repetitive Sequences, Amino Acid ,Patch-Clamp Techniques ,Recombinant Fusion Proteins ,Green Fluorescent Proteins ,Muscle Proteins ,Motility ,In Vitro Techniques ,Motor Endplate ,Membrane Potentials ,Animals, Genetically Modified ,Escape Reaction ,Postsynaptic potential ,Animals ,Receptors, Cholinergic ,Patch clamp ,ARTICLE ,Muscle, Skeletal ,Receptor ,Zebrafish ,Acetylcholine receptor ,Behavior, Animal ,Muscle fatigue ,biology ,General Neuroscience ,Homozygote ,Receptor Aggregation ,biology.organism_classification ,Electric Stimulation ,Luminescent Proteins ,Tetratricopeptide ,Muscle Fatigue ,Synapses ,Neuroscience - Abstract
Upon touch, twitch once zebrafish respond with one or two swimming strokes instead of typical full-blown escapes. This use-dependent fatigue is shown to be a consequence of a mutation in the tetratricopeptide domain of muscle rapsyn, inhibiting formation of subsynaptic acetylcholine receptor clusters. Physiological analysis indicates that reduced synaptic strength, attributable to loss of receptors, is augmented by a potent postsynaptic depression not seen at normal neuromuscular junctions. The synergism between these two physiological processes is causal to the use-dependent muscle fatigue. These findings offer insights into the physiological basis of human myasthenic syndrome and reveal the first demonstration of a role for rapsyn in regulating synaptic function.
- Published
- 2002
46. Author response: Polycomb- and REST-associated histone deacetylases are independent pathways toward a mature neuronal phenotype
- Author
-
Gail Mandel, Huilan Yao, James C. McGann, Saurabh K. Garg, Lujian Liao, Xin Feng, Jun Liu, John R. Yates, and Jon A Oyer
- Subjects
Histone ,biology ,Neuronal phenotype ,biology.protein ,Rest (music) ,Cell biology - Published
- 2014
47. Polycomb- and REST-associated histone deacetylases are independent pathways toward a mature neuronal phenotype
- Author
-
Saurabh K. Garg, James C. McGann, Huilan Yao, Xin Feng, Gail Mandel, Jun Liu, Lujian Liao, John R. Yates, and Jon A Oyer
- Subjects
QH301-705.5 ,Cellular differentiation ,Science ,Repressor ,RE1-silencing transcription factor ,Methylation ,Histone Deacetylases ,General Biochemistry, Genetics and Molecular Biology ,neuronal ,Histones ,Mice ,poised ,Animals ,ES cell ,Biology (General) ,Promoter Regions, Genetic ,Embryonic Stem Cells ,mouse ,Neurons ,Genetics ,General Immunology and Microbiology ,biology ,Lysine ,General Neuroscience ,REST ,Polycomb Repressive Complex 2 ,Cell Differentiation ,General Medicine ,ES cells ,Chromatin ,Repressor Proteins ,Polycomb ,Phenotype ,Developmental Biology and Stem Cells ,Gene Expression Regulation ,Genes and Chromosomes ,histone deacetylase ,biology.protein ,H3K4me3 ,Medicine ,Histone deacetylase activity ,Histone deacetylase ,PRC2 ,Research Article - Abstract
The bivalent hypothesis posits that genes encoding developmental regulators required for early lineage decisions are poised in stem/progenitor cells by the balance between a repressor histone modification (H3K27me3), mediated by the Polycomb Repressor Complex 2 (PRC2), and an activator modification (H3K4me3). In this study, we test whether this mechanism applies equally to genes that are not required until terminal differentiation. We focus on the RE1 Silencing Transcription Factor (REST) because it is expressed highly in stem cells and is an established global repressor of terminal neuronal genes. Elucidation of the REST complex, and comparison of chromatin marks and gene expression levels in control and REST-deficient stem cells, shows that REST target genes are poised by a mechanism independent of Polycomb, even at promoters which bear the H3K27me3 mark. Specifically, genes under REST control are actively repressed in stem cells by a balance of the H3K4me3 mark and a repressor complex that relies on histone deacetylase activity. Thus, chromatin distinctions between pro-neural and terminal neuronal genes are established at the embryonic stem cell stage by two parallel, but distinct, repressor pathways. DOI: http://dx.doi.org/10.7554/eLife.04235.001, eLife digest When an embryo is developing, genes are switched on or off at different times, for many different reasons. Many of these genes are switched off, or repressed, by making the DNA inaccessible to the various proteins and molecules that control gene activity. This is achieved by altering the way that the DNA is packaged into a compacted structure called chromatin. A host of proteins modify the structure of chromatin: it can be made more tightly packaged, which keeps genes switched off; or it can be made more loosely packaged, which allows the genes within to be accessed and switched on. The stem cells in an embryo are able to give rise to many different types of specialized cell. Genes that determine which cell type a stem cell will eventually become are often kept in a so-called ‘poised’ state, and have chromatin modifications that encourage genes to be switch on, as well as modifications that switch genes off. Current thinking is that this poised state allows these genes to be switched on or off rapidly in response to the signals that the cell receives during development. The only known protein complex that causes the chromatin to become more compacted in this poised state is the Polycomb complex. This complex binds to specific regions of DNA and is thought to allow stem cells to remain able to become different cell types by repressing the genes required for adopting a specialized cell fate. However, it is unclear if this poised state also regulates those genes that control the final stages of a cell becoming a specific cell type. McGann et al. investigated genes that are involved in the final stages of a nerve cell's development. These genes are regulated by another protein called REST, which acts to repress the genes in non-neuronal cells. McGann et al. found that the genes that are regulated by REST in embryonic stem cells from mice also have their chromatin modified in two contrasting ways. Some of the modifications are linked to switching genes on, while others are linked to keeping genes switched off. Thus these genes are also in a poised state. However, for these genes, this state is acquired without the activity of the Polycomb complex. The results of McGann et al. show that two similar, but distinct, mechanisms keep the genes required for the early and the late stages of nerve cell development in a poised state. If this poised state aids the development of other cell types (for example muscle or fat cells), uncovering how it is achieved could improve our ability to direct stem cells to develop into specific cell types and tissues. DOI: http://dx.doi.org/10.7554/eLife.04235.002
- Published
- 2014
48. Regulation of Neuronal Traits by a Novel Transcriptional Complex
- Author
-
Corinna Burger, Josh Chenoweth, Elena Battaglioli, Paul Brehm, Mary E. Anderson, Mariko Moniwa, James R. Davie, Nurit Ballas, Michael G. Rosenfeld, Fouad Atouf, William J. Bowers, María Estela Andrés, Gail Mandel, David W. Rose, and Howard J. Federoff
- Subjects
Histone Deacetylase 2 ,RE1-silencing transcription factor ,PC12 Cells ,Sodium Channels ,Mice ,0302 clinical medicine ,Settore BIO/13 - Biologia Applicata ,Chlorocebus aethiops ,Nerve Growth Factor ,Cells, Cultured ,Cerebral Cortex ,Neurons ,0303 health sciences ,NAV1.2 Voltage-Gated Sodium Channel ,biology ,General Neuroscience ,Serine Endopeptidases ,Cell Differentiation ,Zinc Fingers ,Chromatin ,Recombinant Proteins ,Cell biology ,DNA-Binding Proteins ,COS Cells ,Recombinant Fusion Proteins ,Neuroscience(all) ,Repressor ,Nerve Tissue Proteins ,Transfection ,Histone Deacetylases ,Cell Line ,03 medical and health sciences ,Bacterial Proteins ,Animals ,Humans ,Psychological repression ,Rest (music) ,030304 developmental biology ,Sodium channel ,Embryo, Mammalian ,Molecular biology ,Rats ,Mice, Inbred C57BL ,Repressor Proteins ,RCOR1 ,biology.protein ,Corepressor ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
The transcriptional repressor, REST, helps restrict neuronal traits to neurons by blocking their expression in nonneuronal cells. To examine the repercussions of REST expression in neurons, we generated a neuronal cell line that expresses REST conditionally. REST expression inhibited differentiation by nerve growth factor, suppressing both sodium current and neurite growth. A novel corepressor complex, CoREST/HDAC2, was shown to be required for REST repression. In the presence of REST, the CoREST/HDAC2 complex occupied the native Nav1.2 sodium channel gene in chromatin. In neuronal cells that lack REST and express sodium channels, the corepressor complex was not present on the gene. Collectively, these studies define a novel HDAC complex that is recruited by the C-terminal repressor domain of REST to actively repress genes essential to the neuronal phenotype.
- Published
- 2001
- Full Text
- View/download PDF
49. Compact Myelin Dictates the Differential Targeting of Two Sodium Channel Isoforms in the Same Axon
- Author
-
Gary Matthews, James S. Trimmer, Tatiana Boiko, S. Rock Levinson, John H. Caldwell, Matthew N. Rasband, and Gail Mandel
- Subjects
Retinal Ganglion Cells ,Gene isoform ,Neuroscience(all) ,Sodium Channels ,Rats, Sprague-Dawley ,Mice ,Mice, Neurologic Mutants ,Compact myelin ,Paranodal junction ,Ranvier's Nodes ,medicine ,Animals ,Protein Isoforms ,Peripheral Nerves ,Axon ,Myelin Sheath ,Chemistry ,General Neuroscience ,Sodium channel ,Optic Nerve ,Immunohistochemistry ,Axons ,Rats ,Cell biology ,Sprague dawley ,medicine.anatomical_structure ,nervous system ,Neuroscience - Abstract
Voltage-dependent sodium channels are uniformly distributed along unmyelinated axons, but are highly concentrated at nodes of Ranvier in myelinated axons. Here, we show that this pattern is associated with differential localization of distinct sodium channel alpha subunits to the unmyelinated and myelinated zones of the same retinal ganglion cell axons. In adult axons, Na(v)1.2 is localized to the unmyelinated zone, whereas Na(v)1.6 is specifically targeted to nodes. During development, Na(v)1.2 is expressed first and becomes clustered at immature nodes of Ranvier, but as myelination proceeds, Na(v)1.6 replaces Na(v)1.2 at nodes. In Shiverer mice, which lack compact myelin, Na(v)1.2 is found throughout adult axons, whereas little Na(v)1.6 is detected. Together, these data show that sodium channel isoforms are differentially targeted to distinct domains of the same axon in a process associated with formation of compact myelin.
- Published
- 2001
50. Progesterone treatment abolishes exogenously expressed ionic currents inXenopusoocytes
- Author
-
Christopher M. Davenport, Anatoly Shcherbatko, Simon R. Levinson, Paul Brehm, Gail Mandel, and Joan C. Speh
- Subjects
Cytoplasm ,medicine.medical_specialty ,Potassium Channels ,Physiology ,Xenopus ,medicine.medical_treatment ,Down-Regulation ,Inositol 1,4,5-Trisphosphate ,Sodium Channels ,Meiosis ,Salientia ,Internal medicine ,CDC2 Protein Kinase ,Gene expression ,Cyclic AMP ,Potassium Channel Blockers ,medicine ,Animals ,Cytoskeleton ,Progesterone ,biology ,Sodium channel ,Cell Membrane ,Electric Conductivity ,Proteins ,Cell Biology ,biology.organism_classification ,Oocyte ,Cell biology ,Steroid hormone ,medicine.anatomical_structure ,Endocrinology ,Oocytes ,Female ,Sodium Channel Blockers - Abstract
Fully grown oocytes of Xenopus laevis undergo resumption of the meiotic cycle when treated with the steroid hormone progesterone. Previous studies have shown that meiotic maturation results in profound downregulation of specific endogenous membrane proteins in oocytes. To determine whether the maturation impacts the functional properties of exogenously expressed membrane proteins, we used cut-open recordings from Xenopus oocytes expressing several types of Na+and K+channels. Treatment of oocytes with progesterone resulted in a downregulation of heterologously expressed Na+and K+channels without a change in the kinetics of the currents. The time course of progesterone-induced ion channel inhibition was concentration dependent. Complete elimination of Na+currents temporally coincided with development of germinal vesicle breakdown, while elimination of K+currents was delayed by ∼2 h. Coexpression of human β1-subunit with rat skeletal muscle α-subunit in Xenopus oocytes did not prevent progesterone-induced downregulation of Na+channels. Addition of 8-bromo-cAMP to oocytes or injection of heparin before progesterone treatment prevented the loss of expressed currents. Pharmacological studies suggest that the inhibitory effects of progesterone on expressed Na+and K+channels occur downstream of the activation of cdc2 kinase. The loss of channels is correlated with a reduction in Na+channel immunofluorescence, pointing to a disappearance of the ion channel-forming proteins from the surface membrane.
- Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.