455 results on '"Guo-Li Ming"'
Search Results
152. Tet3 regulates synaptic transmission and homeostatic plasticity via DNA oxidation and repair
- Author
-
Guo Li Ming, Chun Zhong, Daniel H. Geschwind, Huimei Yu, Junjie U. Guo, Jaehoon Shin, Fuying Gao, Yi Lan Weng, Giovanni Coppola, Hongjun Song, and Yijing Su
- Subjects
DNA Repair ,Nonsynaptic plasticity ,Biology ,In Vitro Techniques ,Inbred C57BL ,Synaptic Transmission ,Article ,Dioxygenases ,Mice ,Cellular neuroscience ,Homeostatic plasticity ,Proto-Oncogene Proteins ,Metaplasticity ,Receptors ,AMPA ,Psychology ,Animals ,Homeostasis ,Receptors, AMPA ,Synaptic scaling ,Neurology & Neurosurgery ,Neuronal Plasticity ,General Neuroscience ,Neurosciences ,DNA ,DNA Methylation ,Cell biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Synaptic fatigue ,DNA demethylation ,Gene Knockdown Techniques ,Synaptic plasticity ,Cognitive Sciences ,Neuroscience ,Oxidation-Reduction ,Signal Transduction - Abstract
Contrary to the long-held belief that DNA methylation of terminally differentiated cells is permanent and essentially immutable, post-mitotic neurons exhibit extensive DNA demethylation. The cellular function of active DNA demethylation in neurons, however, remains largely unknown. Tet family proteins oxidize 5-methylcytosine to initiate active DNA demethylation through the base-excision repair pathway. Here, we show that synaptic activity bi-directionally regulates neuronal Tet3 expression. Functionally, knockdown of Tet or inhibition of base-excision repair in hippocampal neurons elevates excitatory glutamatergic synaptic transmission, whereas overexpressing Tet3 or Tet1 catalytic domain decreases it. Furthermore, dysregulation of Tet3 signalling prevents homeostatic synaptic plasticity. Mechanistically, Tet3 dictates neuronal surface GluR1 levels. RNA-seq analyses further revealed a pivotal role of Tet3 in regulating gene expression in response to global synaptic activity changes. Thus, Tet3 serves as a synaptic activity sensor to epigenetically regulate fundamental properties and meta-plasticity of neurons via active DNA demethylation.
- Published
- 2015
153. In vivo clonal analysis reveals spatiotemporal regulation of thalamic nucleogenesis
- Author
-
Guo Li Ming, Wenhui Mu, Earl Parker Scott, Hongjun Song, Madeline Freeman, Qing-Feng Wu, Ella Borgenheimer, Yasushi Nakagawa, Samuel Z.H. Wong, and Xize Guo
- Subjects
0301 basic medicine ,Cell division ,Cellular differentiation ,Clone (cell biology) ,Regulator ,Mice ,0302 clinical medicine ,Thalamus ,Neural Stem Cells ,Animal Cells ,Genes, Reporter ,Pregnancy ,Gene expression ,Medicine and Health Sciences ,Basic Helix-Loop-Helix Transcription Factors ,Cell Cycle and Cell Division ,Biology (General) ,Cell Analysis ,Neurons ,Regulation of gene expression ,0303 health sciences ,Stem Cells ,General Neuroscience ,Neurogenesis ,Brain ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Bioassays and Physiological Analysis ,Cell Division Analysis ,Cell Processes ,Cell Tracking ,Thalamic Nuclei ,Female ,Anatomy ,Cellular Types ,Stem cell ,General Agricultural and Biological Sciences ,Cell Division ,Research Article ,QH301-705.5 ,Period (gene) ,Green Fluorescent Proteins ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Research and Analysis Methods ,Zinc Finger Protein GLI1 ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Developmental Neuroscience ,Animals ,Cell Lineage ,Progenitor cell ,Molecular Biology Techniques ,Molecular Biology ,030304 developmental biology ,Integrases ,General Immunology and Microbiology ,Embryogenesis ,Biology and Life Sciences ,Cell Biology ,Embryo, Mammalian ,Clone Cells ,030104 developmental biology ,Cellular Neuroscience ,Neuroscience ,030217 neurology & neurosurgery ,Cloning - Abstract
The thalamus, a crucial regulator of cortical functions, is composed of many nuclei arranged in a spatially complex pattern. Thalamic neurogenesis occurs over a short period during mammalian embryonic development. These features have hampered the effort to understand how regionalization, cell divisions, and fate specification are coordinated and produce a wide array of nuclei that exhibit distinct patterns of gene expression and functions. Here, we performed in vivo clonal analysis to track the divisions of individual progenitor cells and spatial allocation of their progeny in the developing mouse thalamus. Quantitative analysis of clone compositions revealed evidence for sequential generation of distinct sets of thalamic nuclei based on the location of the founder progenitor cells. Furthermore, we identified intermediate progenitor cells that produced neurons populating more than one thalamic nuclei, indicating a prolonged specification of nuclear fate. Our study reveals an organizational principle that governs the spatial and temporal progression of cell divisions and fate specification and provides a framework for studying cellular heterogeneity and connectivity in the mammalian thalamus., Author summary The thalamus—a brain structure commonly associated with relaying sensory information between cortex and other regions—is organized into many cell clusters called nuclei. Each thalamic nucleus is populated by neurons with distinct patterns of gene expression and connections to other brain regions and plays a distinct role in cortical functions. In this study, we performed an analysis of developing cells in the thalamus, using the mosaic analysis with double markers (MADM) method in mice, a technique that allows the labeling of descendants of dividing cells. Using 3 different transgenic mouse lines allowed us to determine the cell lineage of thalamic progenitor cells at different locations and stages of differentiation. By genetically labeling single progenitor cells, we measured how cell division and maturation occurs during the brief time span when neurons are generated. Our data also show how neurons eventually contribute to multiple nuclei across the thalamus. The organizational principles that we found in the thalamus might apply to the development of other brain structures that are composed of multiple nuclei.
- Published
- 2017
- Full Text
- View/download PDF
154. DISC1 Regulates Neurogenesis via Modulating Kinetochore Attachment of Ndel1/Nde1 during Mitosis
- Author
-
Eunchai Kang, Guo Li Ming, Jieun Kim, Mao Mao, Cong Yu, Xuyu Qian, Mingjie Zhang, Fadi Jacob, Fei Ye, Chuan Yu, Hongjun Song, and Randy Yat Choi Poon
- Subjects
0301 basic medicine ,Male ,Models, Molecular ,Pluripotent Stem Cells ,Neurogenesis ,Mitosis ,Nerve Tissue Proteins ,Article ,03 medical and health sciences ,DISC1 ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Pregnancy ,medicine ,Animals ,Humans ,Induced pluripotent stem cell ,Neurons ,NDEL1 ,biology ,Kinetochore ,General Neuroscience ,Cell Cycle ,Cell cycle ,Embryonic stem cell ,Immunohistochemistry ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Schizophrenia ,Female ,Neuron ,Carrier Proteins ,030217 neurology & neurosurgery ,Centrosome localization ,HeLa Cells ,Protein Binding - Abstract
Summary Mutations of DISC1 (disrupted-in-schizophrenia 1) have been associated with major psychiatric disorders. Despite the hundreds of DISC1-binding proteins reported, almost nothing is known about how DISC1 interacts with other proteins structurally to impact human brain development. Here we solved the high-resolution structure of DISC1 C-terminal tail in complex with its binding domain of Ndel1. Mechanistically, DISC1 regulates Ndel1's kinetochore attachment, but not its centrosome localization, during mitosis. Functionally, disrupting DISC1/Ndel1 complex formation prolongs mitotic length and interferes with cell-cycle progression in human cells, and it causes cell-cycle deficits of radial glial cells in the embryonic mouse cortex and human forebrain organoids. We also observed similar deficits in organoids derived from schizophrenia patient induced pluripotent stem cells (iPSCs) with a DISC1 mutation that disrupts its interaction with Ndel1. Our study uncovers a new mechanism of action for DISC1 based on its structure, and it has implications for how genetic insults may contribute to psychiatric disorders.
- Published
- 2017
155. Animal and Cellular Models of Psychotic Disorders
- Author
-
Mikhail V. Pletnikov, Guo-Li Ming, and Christopher A. Ross
- Abstract
Animal and cell models are experimental systems developed to study particular aspects of a disease, as no model can accurately reflect all features of the disease. In this critical review we mention some of the nongenetic models but focus on genetic mouse models, evaluate their advantages and limitations, and comment on potential new prospects for the field. The ability to reprogram somatic cells from patients and unaffected donors to induced pluripotent stem cells (iPSCs) has the potential to substantially enhance our knowledge of normal cellular development and disease pathogenesis. The use of cell and animal models will help elucidate basic cellular and molecular mechanisms of pathogenesis, which will enable the development of targeted therapeutic approaches.
- Published
- 2017
- Full Text
- View/download PDF
156. m
- Author
-
Hailing, Shi, Xuliang, Zhang, Yi-Lan, Weng, Zongyang, Lu, Yajing, Liu, Zhike, Lu, Jianan, Li, Piliang, Hao, Yu, Zhang, Feng, Zhang, You, Wu, Jary Y, Delgado, Yijing, Su, Meera J, Patel, Xiaohua, Cao, Bin, Shen, Xingxu, Huang, Guo-Li, Ming, Xiaoxi, Zhuang, Hongjun, Song, Chuan, He, and Tao, Zhou
- Subjects
Male ,Mice, Knockout ,Neurons ,Binding Sites ,Neuronal Plasticity ,Adenine ,Spatial Learning ,RNA-Binding Proteins ,Methyltransferases ,Hippocampus ,Synaptic Transmission ,Article ,Mice ,Memory ,Protein Biosynthesis ,Animals ,Female ,RNA, Messenger - Abstract
Summary N6-methyladenosine (m6A), the most prevalent internal RNA modification on mammalian messenger RNAs (mRNAs), regulates fates and functions of modified transcripts through m6A-specific binding proteins1–5. m6A is abundant in the nervous system and modulates various neural functions6–11. While m6A marks groups of mRNAs for coordinated degradation in various physiological processes12–15, the relevance of m6A in mRNA translation remains largely unknown in vivo. Here we show that, through its binding protein Ythdf1, m6A promotes protein synthesis of target transcripts in response to neuronal stimuli in the adult mouse hippocampus, thereby facilitating learning and memory. Mice with genetic deletion of Ythdf1 (Ythdf1-KO) exhibit learning and memory defects as well as impaired hippocampal synaptic transmission and long-term potentiation. Ythdf1 re-expression in the hippocampus of adult Ythdf1-KO mice rescues behavioral and synaptic defects, while hippocampus-specific acute knockdown of Ythdf1 or Mettl3, the catalytic component of m6A methyltransferase complex, recapitulates the hippocampal deficiency. Transcriptome-wide mapping of Ythdf1 binding sites and m6A sites on hippocampal mRNAs uncovered key neuronal genes. Nascent protein labeling and tether reporter assays in hippocampal neurons revealed that Ythdf1 enhances protein synthesis in a neuronal-stimulus-dependent manner. Collectively, our results uncover a pathway of mRNA m6A methylation in learning and memory, which is mediated through Ythdf1 in response to stimuli.
- Published
- 2017
157. Multiplexed Biomarker Panels Discriminate Zika and Dengue Virus Infection in Humans
- Author
-
Jorge L. Muñoz-Jordán, Hengli Tang, Guo Li Ming, Ki Jun Yoon, Jianbo Pan, Heng Zhu, Hongjun Song, Daniel Eichinger, Freddy A. Medina, Pedro Ramos, Hee Sool Rho, Jiang Qian, Ignacio Pino, Guang Song, and Emily M. Lee
- Subjects
0301 basic medicine ,DNA, Complementary ,viruses ,Protein Array Analysis ,Dengue virus ,medicine.disease_cause ,Biochemistry ,Analytical Chemistry ,Dengue fever ,Zika virus ,Dengue ,03 medical and health sciences ,Viral Proteins ,0302 clinical medicine ,Blood serum ,medicine ,Humans ,Molecular Biology ,NS3 ,biology ,business.industry ,Zika Virus Infection ,Research ,Outbreak ,virus diseases ,Zika Virus ,biochemical phenomena, metabolism, and nutrition ,Dengue Virus ,medicine.disease ,biology.organism_classification ,Virology ,030104 developmental biology ,Capsid ,Immunoglobulin M ,030220 oncology & carcinogenesis ,Immunology ,DNA, Viral ,Biomarker (medicine) ,business ,Biomarkers - Abstract
Zika virus (ZIKV) and dengue virus (DENV) are closely related flaviviruses that cause widespread, acute febrile illnesses, notably microcephaly for fetuses of infected pregnant women. Detecting the viral cause of these illnesses is paramount to determine risks to patients, counsel pregnant women, and help fight outbreaks. A combined diagnostic algorithm for ZIKV and DENV requires Reverse transcription polymerase chain reaction (RT-PCR) and IgM antibody detection. RT-PCR differentiates between DENV and ZIKV infections during the acute phases of infection, but differentiation based on IgM antibodies is currently nearly impossible in endemic areas. We have developed a ZIKV/DENV protein array and tested it with serum samples collected from ZIKV- and DENV-infected patients and healthy subjects in Puerto Rico. Our analyses reveal a biomarker panel that are capable of discriminating ZIKV and DENV infections with high accuracy, including Capsid protein from African ZIKV strain MR766, and other 5 pair of proteins (NS1, NS2A, NS3, NS4B and NS5) from ZIKV and DENV respectively. Both sensitivity and specificity of the test for ZIKV from DENV are around 90%. We propose that the ZIKV/DENV protein array will be used in future studies to discriminate patients infected with ZIKV from DENV.
- Published
- 2017
158. Epitranscriptomic m
- Author
-
Yi-Lan, Weng, Xu, Wang, Ran, An, Jessica, Cassin, Caroline, Vissers, Yuanyuan, Liu, Yajing, Liu, Tianlei, Xu, Xinyuan, Wang, Samuel Zheng Hao, Wong, Jessica, Joseph, Louis C, Dore, Qiang, Dong, Wei, Zheng, Peng, Jin, Hao, Wu, Bin, Shen, Xiaoxi, Zhuang, Chuan, He, Kai, Liu, Hongjun, Song, and Guo-Li, Ming
- Subjects
Mice, Knockout ,Adenosine ,Sensory Receptor Cells ,Transcription, Genetic ,Nerve Crush ,PTEN Phosphohydrolase ,RNA-Binding Proteins ,Nerve Tissue Proteins ,Methyltransferases ,Sciatic Nerve ,Axons ,Epigenesis, Genetic ,Nerve Regeneration ,Gene Ontology ,Ganglia, Spinal ,Animals ,RNA, Messenger ,RNA Processing, Post-Transcriptional ,Sciatic Neuropathy - Abstract
N
- Published
- 2017
159. Temporal Control of Mammalian Cortical Neurogenesis by m
- Author
-
Ki-Jun, Yoon, Francisca Rojas, Ringeling, Caroline, Vissers, Fadi, Jacob, Michael, Pokrass, Dennisse, Jimenez-Cyrus, Yijing, Su, Nam-Shik, Kim, Yunhua, Zhu, Lily, Zheng, Sunghan, Kim, Xinyuan, Wang, Louis C, Doré, Peng, Jin, Sergi, Regot, Xiaoxi, Zhuang, Stefan, Canzar, Chuan, He, Guo-Li, Ming, and Hongjun, Song
- Subjects
Mice, Knockout ,Neurogenesis ,RNA Stability ,Cell Cycle ,Gene Expression Regulation, Developmental ,Methyltransferases ,Methylation ,Article ,Organoids ,Mice ,Prosencephalon ,Gene Expression Regulation ,Neural Stem Cells ,Gene Knockdown Techniques ,Animals ,Humans ,RNA, Messenger ,RNA Processing, Post-Transcriptional - Abstract
Modification of messenger RNAs through a process called m6A methylation facilitates dynamic temporal regulation of RNA levels in neural precursor cells, enabling fine-tuning of developing neuronal circuits in the brain.
- Published
- 2017
160. Methylated cis-regulatory elements mediate KLF4-dependent gene transactivation and cell migration
- Author
-
Guo Li Ming, Olutobi Oyinlade, Jun Wan, Shuli Xia, Jiang Qian, Sheng Liu, Brian Tung, Mingyao Ying, Yijing Su, Hongjun Song, Qifeng Song, and Heng Zhu
- Subjects
Transcriptional Activation ,0301 basic medicine ,cell migration ,QH301-705.5 ,Science ,Kruppel-Like Transcription Factors ,Regulatory Sequences, Nucleic Acid ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,Kruppel-Like Factor 4 ,03 medical and health sciences ,Epigenetics of physical exercise ,Cell Movement ,Cell Line, Tumor ,Histone methylation ,Humans ,histone modification ,Biology (General) ,RNA-Directed DNA Methylation ,Epigenomics ,Genetics ,DNA methylation ,General Immunology and Microbiology ,General Neuroscience ,Pioneer factor ,Cell Biology ,General Medicine ,Chromatin Assembly and Disassembly ,KLF4 ,Chromatin ,030104 developmental biology ,Gene Expression Regulation ,Genes and Chromosomes ,Medicine ,Research Article ,Human ,Protein Binding - Abstract
Altered DNA methylation status is associated with human diseases and cancer; however, the underlying molecular mechanisms remain elusive. We previously identified many human transcription factors, including Krüppel-like factor 4 (KLF4), as sequence-specific DNA methylation readers that preferentially recognize methylated CpG (mCpG), here we report the biological function of mCpG-dependent gene regulation by KLF4 in glioblastoma cells. We show that KLF4 promotes cell adhesion, migration, and morphological changes, all of which are abolished by R458A mutation. Surprisingly, 116 genes are directly activated via mCpG-dependent KLF4 binding activity. In-depth mechanistic studies reveal that recruitment of KLF4 to the methylated cis-regulatory elements of these genes result in chromatin remodeling and transcription activation. Our study demonstrates a new paradigm of DNA methylation-mediated gene activation and chromatin remodeling, and provides a general framework to dissect the biological functions of DNA methylation readers and effectors. DOI: http://dx.doi.org/10.7554/eLife.20068.001
- Published
- 2017
161. How does Zika virus cause microcephaly?
- Author
-
Guo Li Ming, Zhexing Wen, and Hongjun Song
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Microcephaly ,Review ,Biology ,World health ,Zika virus ,Disease Outbreaks ,03 medical and health sciences ,Genetics ,medicine ,ZikV Infection ,Animals ,Humans ,Zika Virus Infection ,Public health ,Outbreak ,Zika Virus ,Mammalian brain ,biology.organism_classification ,medicine.disease ,Virology ,Flavivirus ,030104 developmental biology ,Developmental Biology - Abstract
The re-emergence of Zika virus (ZIKV), a mosquito-borne and sexually transmitted flavivirus circulating in >70 countries and territories, poses a significant global threat to public health due to its ability to cause severe developmental defects in the human brain, such as microcephaly. Since the World Health Organization declared the ZIKV outbreak a Public Health Emergency of International Concern, remarkable progress has been made to gain insight into cellular targets, pathogenesis, and underlying biological mechanisms of ZIKV infection. Here we review the current knowledge and progress in understanding the impact of ZIKV exposure on the mammalian brain development and discuss potential underlying mechanisms.
- Published
- 2017
162. An intrinsic epigenetic barrier for functional axon regeneration
- Author
-
Jessica Cassin, Xinzhong Dong, Seung Gi Jin, Ahmet Hoke, Jessica Joseph, Yi Lan Weng, Gerd P. Pfeifer, Guo Li Ming, Ran An, Ruifa Mi, Hongjun Song, Chun Zhong, Zhigang He, Alfonso Bellacosa, and Chen Wang
- Subjects
0301 basic medicine ,Biology ,Retinal ganglion ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,Dorsal root ganglion ,Peripheral Nerve Injuries ,Ganglia, Spinal ,medicine ,Animals ,Axon ,General Neuroscience ,Regeneration (biology) ,Axons ,Nerve Regeneration ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,DNA demethylation ,nervous system ,Peripheral nervous system ,Peripheral nerve injury ,5-Methylcytosine ,Sciatic nerve ,Neuroscience - Abstract
Mature neurons in the adult peripheral nervous system can effectively switch from a dormant state with little axonal growth to robust axon regeneration upon injury. The mechanisms by which injury unlocks mature neurons’ intrinsic axonal growth competence are not well understood. Here we show that peripheral sciatic nerve lesion in adult mice leads to elevated levels of Tet3 and 5- hydroxylmethylcytosine in dorsal root ganglion (DRG) neurons. Functionally, Tet3 is required for robust axon regeneration of DRG neurons and behavioral recovery. Mechanistically, peripheral nerve injury induces DNA demethylation and upregulation of multiple regeneration-associated genes in a Tet3- and thymine DNA glycosylase-dependent fashion in DRG neurons. In addition, Pten deletion-induced axon regeneration of retinal ganglion neurons in the adult central nervous system is attenuated upon Tet1 knockdown. Together, our study suggests an epigenetic barrier that can be removed by active DNA demethylation to permit axon regeneration in the adult mammalian nervous system.
- Published
- 2017
163. Author response: Methylated cis-regulatory elements mediate KLF4-dependent gene transactivation and cell migration
- Author
-
Jiang Qian, Shuli Xia, Qifeng Song, Heng Zhu, Mingyao Ying, Sheng Liu, Brian Tung, Hongjun Song, Guo Li Ming, Jun Wan, Yijing Su, and Olutobi Oyinlade
- Subjects
Transactivation ,KLF4 ,Cell migration ,Biology ,Gene ,Cis-regulatory element ,Cell biology - Published
- 2017
- Full Text
- View/download PDF
164. A Patient-Derived Glioblastoma Organoid Model and Biobank Recapitulates Inter- and Intra-tumoral Heterogeneity
- Author
-
Dmitriy Petrov, Di-ao Liu, H. Isaac Chen, Deeksha Saxena, Phuong T.T. Nguyen, Xuyu Qian, MacLean Nasrallah, Radhika Thokala, Samuel Zheng Hao Wong, Timothy H. Lucas, Jay F. Dorsey, Fadi Jacob, Donald M. O'Rourke, Steven Brem, Ryan D Salinas, Kimberly M. Christian, Guo Li Ming, Hongjun Song, Jordan G. Schnoll, Daniel Y. Zhang, Stefan Prokop, Saad Sheikh, and Zev A. Binder
- Subjects
Adult ,Male ,medicine.medical_treatment ,Cell Culture Techniques ,Mice, Nude ,Biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Organoid ,Animals ,Humans ,Aged ,Biological Specimen Banks ,030304 developmental biology ,Aged, 80 and over ,0303 health sciences ,Translational bioinformatics ,Reproducibility of Results ,Immunotherapy ,Middle Aged ,medicine.disease ,Key features ,Xenograft Model Antitumor Assays ,Biobank ,Chimeric antigen receptor ,Organoids ,Cancer research ,Treatment strategy ,Female ,Glioblastoma ,030217 neurology & neurosurgery - Abstract
Summary Glioblastomas exhibit vast inter- and intra-tumoral heterogeneity, complicating the development of effective therapeutic strategies. Current in vitro models are limited in preserving the cellular and mutational diversity of parental tumors and require a prolonged generation time. Here, we report methods for generating and biobanking patient-derived glioblastoma organoids (GBOs) that recapitulate the histological features, cellular diversity, gene expression, and mutational profiles of their corresponding parental tumors. GBOs can be generated quickly with high reliability and exhibit rapid, aggressive infiltration when transplanted into adult rodent brains. We further demonstrate the utility of GBOs to test personalized therapies by correlating GBO mutational profiles with responses to specific drugs and by modeling chimeric antigen receptor T cell immunotherapy. Our studies show that GBOs maintain many key features of glioblastomas and can be rapidly deployed to investigate patient-specific treatment strategies. Additionally, our live biobank establishes a rich resource for basic and translational glioblastoma research.
- Published
- 2020
- Full Text
- View/download PDF
165. m6A mRNA Methylation Is Essential for Oligodendrocyte Maturation and CNS Myelination
- Author
-
Yang I. Li, Qili Fei, Rejani B. Kunjamma, Yulia Dzhashiashvili, Joshua S. Jones, Benayahu Elbaz, Xiaoxi Zhuang, Huan Xu, Brian Popko, Yi Lan Weng, Guo Li Ming, Chuan He, and Ankeeta Shah
- Subjects
0301 basic medicine ,CNS hypomyelination ,Messenger RNA ,RNA methylation ,General Neuroscience ,RNA ,Biology ,Oligodendrocyte ,Cell biology ,Transcriptome ,03 medical and health sciences ,Myelin ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,MRNA methylation ,030217 neurology & neurosurgery - Abstract
Summary The molecular mechanisms that govern the maturation of oligodendrocyte lineage cells remain unclear. Emerging studies have shown that N6-methyladenosine (m6A), the most common internal RNA modification of mammalian mRNA, plays a critical role in various developmental processes. Here, we demonstrate that oligodendrocyte lineage progression is accompanied by dynamic changes in m6A modification on numerous transcripts. In vivo conditional inactivation of an essential m6A writer component, METTL14, results in decreased oligodendrocyte numbers and CNS hypomyelination, although oligodendrocyte precursor cell (OPC) numbers are normal. In vitro Mettl14 ablation disrupts postmitotic oligodendrocyte maturation and has distinct effects on OPC and oligodendrocyte transcriptomes. Moreover, the loss of Mettl14 in oligodendrocyte lineage cells causes aberrant splicing of myriad RNA transcripts, including those that encode the essential paranodal component neurofascin 155 (NF155). Together, our findings indicate that dynamic RNA methylation plays an important regulatory role in oligodendrocyte development and CNS myelination.
- Published
- 2020
- Full Text
- View/download PDF
166. Modeling non-syndromic autism and the impact of TRPC6 disruption in human neurons
- Author
-
Michael F. Walker, Xiang Q. Gu, Stephen Sanders, Allan Acab, Nicholas C. Spitzer, Dhiraj K. Pradhan, Daniele Yumi Sunaga, Guo Li Ming, Richard P. Lifton, Alysson R. Muotri, Gabriel G. Haddad, Matthew W. State, Weizhen Ji, Karina Griesi-Oliveira, Estevão Vadasz, Hongjun Song, Yanelli Nunez, Maria C. Marchetto, Abha R. Gupta, Maria Rita Passos-Bueno, Xavier Nicol, Thanathom Chailangkarn, Thomas V. Fernandez, John D. Murdoch, and Alexander Dietrich
- Subjects
Male ,medicine.disease_cause ,Carboplatin ,Mice ,Antineoplastic Combined Chemotherapy Protocols ,disease modeling ,Child ,Cells, Cultured ,Etoposide ,Neurons ,education.field_of_study ,Mutation ,Cell Differentiation ,Penetrance ,Psychiatry and Mental health ,Haploinsufficiency ,Signal Transduction ,induced pluripotent stem cells ,Prednisolone ,Population ,autism ,Mice, Transgenic ,Rett syndrome ,In Vitro Techniques ,Biology ,Article ,Cell Line ,MECP2 ,Cellular and Molecular Neuroscience ,mental disorders ,TRPC6 Cation Channel ,medicine ,Animals ,Humans ,Autistic Disorder ,education ,Molecular Biology ,Methyl-CpG binding ,Cell Proliferation ,TRPC Cation Channels ,Embryo, Mammalian ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,Gene Expression Regulation ,Inhibitory Postsynaptic Potentials ,Autism ,Mitoxantrone ,Neuroscience - Abstract
An increasing number of genetic variants have been implicated in autism spectrum disorders (ASDs), and the functional study of such variants will be critical for the elucidation of autism pathophysiology. Here, we report a de novo balanced translocation disruption of TRPC6, a cation channel, in a non-syndromic autistic individual. Using multiple models, such as dental pulp cells, induced pluripotent stem cell (iPSC)-derived neuronal cells and mouse models, we demonstrate that TRPC6 reduction or haploinsufficiency leads to altered neuronal development, morphology and function. The observed neuronal phenotypes could then be rescued by TRPC6 complementation and by treatment with insulin-like growth factor-1 or hyperforin, a TRPC6-specific agonist, suggesting that ASD individuals with alterations in this pathway may benefit from these drugs. We also demonstrate that methyl CpG binding protein-2 (MeCP2) levels affect TRPC6 expression. Mutations in MeCP2 cause Rett syndrome, revealing common pathways among ASDs. Genetic sequencing of TRPC6 in 1041 ASD individuals and 2872 controls revealed significantly more nonsynonymous mutations in the ASD population, and identified loss-of-function mutations with incomplete penetrance in two patients. Taken together, these findings suggest that TRPC6 is a novel predisposing gene for ASD that may act in a multiple-hit model. This is the first study to use iPSC-derived human neurons to model non-syndromic ASD and illustrate the potential of modeling genetically complex sporadic diseases using such cells.
- Published
- 2014
- Full Text
- View/download PDF
167. Functions and Dysfunctions of Adult Hippocampal Neurogenesis
- Author
-
Kimberly M. Christian, Hongjun Song, and Guo Li Ming
- Subjects
Neurogenesis ,Models, Neurological ,Central nervous system ,Hippocampus ,Hippocampal formation ,Article ,Cognition ,Neural Pathways ,medicine ,Animals ,Humans ,Neurons ,Brain Diseases ,Neuronal Plasticity ,Mechanism (biology) ,General Neuroscience ,Dentate gyrus ,Neural stem cell ,Affect ,Neuropoiesis ,medicine.anatomical_structure ,nervous system ,Psychology ,Neuroscience - Abstract
Adult neurogenesis, a developmental process of generating functionally integrated neurons, occurs throughout life in the hippocampus of the mammalian brain and showcases the highly plastic nature of the mature central nervous system. Significant progress has been made in recent years to decipher how adult neurogenesis contributes to brain functions. Here we review recent findings that inform our understanding of adult hippocampal neurogenesis processes and special properties of adult-born neurons. We further discuss potential roles of adult-born neurons at the circuitry and behavioral levels in cognitive and affective functions and how their dysfunction may contribute to various brain disorders. We end by considering a general model proposing that adult neurogenesis is not a cell-replacement mechanism, but instead maintains a plastic hippocampal neuronal circuit via the continuous addition of immature, new neurons with unique properties and structural plasticity of mature neurons induced by new-neuron integration.
- Published
- 2014
- Full Text
- View/download PDF
168. Modeling a Genetic Risk for Schizophrenia in iPSCs and Mice Reveals Neural Stem Cell Deficits Associated with Adherens Junctions and Polarity
- Author
-
Joel E. Kleinman, Dan Rujescu, Maxwell Towe, Namshik Kim, Joo Heon Shin, Gianluca Ursini, Eva Pekle, Gregory L. Krauss, Kimberly M. Christian, Georgia Makri, Syed Mohammed Qasim Hussaini, Thomas M. Hyde, Guo Li Ming, Hongjun Song, David W. Nauen, Ki-Jun Yun, Yohan Lee, Fengyu Zhang, Youngbin Park, Ce Zhang, Zhexing Wen, Judith L. Rapoport, Raeeun Chung, Daniel R. Weinberger, Ha Nam Nguyen, and David St Clair
- Subjects
Adult ,Risk ,Male ,DNA Copy Number Variations ,Polarity (physics) ,Cell ,Induced Pluripotent Stem Cells ,Mice, Inbred Strains ,Haploinsufficiency ,Biology ,White People ,Article ,Cell Line ,Adherens junction ,Mice ,Mediator ,Neural Stem Cells ,Intellectual Disability ,medicine ,Genetics ,Animals ,Humans ,Copy-number variation ,Genetic risk ,Autistic Disorder ,Progenitor cell ,Induced pluripotent stem cell ,Genetic Association Studies ,Adaptor Proteins, Signal Transducing ,Chromosome Aberrations ,Chromosomes, Human, Pair 15 ,Cell Polarity ,Epistasis, Genetic ,Adherens Junctions ,Cell Biology ,Middle Aged ,medicine.disease ,Neural stem cell ,Wiskott-Aldrich Syndrome Protein Family ,Cell biology ,medicine.anatomical_structure ,Schizophrenia ,Actin-Related Protein 2 ,Molecular Medicine ,Stem cell ,Neuroscience - Abstract
SummaryDefects in brain development are believed to contribute toward the onset of neuropsychiatric disorders, but identifying specific underlying mechanisms has proven difficult. Here, we took a multifaceted approach to investigate why 15q11.2 copy number variants are prominent risk factors for schizophrenia and autism. First, we show that human iPSC-derived neural progenitors carrying 15q11.2 microdeletion exhibit deficits in adherens junctions and apical polarity. This results from haploinsufficiency of CYFIP1, a gene within 15q11.2 that encodes a subunit of the WAVE complex, which regulates cytoskeletal dynamics. In developing mouse cortex, deficiency in CYFIP1 and WAVE signaling similarly affects radial glial cells, leading to their ectopic localization outside of the ventricular zone. Finally, targeted human genetic association analyses revealed an epistatic interaction between CYFIP1 and WAVE signaling mediator ACTR2 and risk for schizophrenia. Our findings provide insight into how CYFIP1 regulates neural stem cell function and may contribute to the susceptibility of neuropsychiatric disorders.
- Published
- 2014
- Full Text
- View/download PDF
169. A previously undetected pathology of Zika virus infection
- Author
-
Guo Li Ming, Kimberly M. Christian, and Hongjun Song
- Subjects
0301 basic medicine ,Cellular pathology ,Offspring ,Neurogenesis ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Zika virus ,03 medical and health sciences ,Fetus ,Pregnancy ,Animals ,Humans ,Pregnancy Complications, Infectious ,Zika Virus Infection ,Zika Virus ,General Medicine ,biology.organism_classification ,Virology ,Nonhuman primate ,Disease Models, Animal ,030104 developmental biology ,Microcephaly ,Female ,Macaca nemestrina ,Neural development ,Neurocognitive - Abstract
Zika virus (ZIKV) is a flavivirus with teratogenic effects on fetal brain, but the spectrum of ZIKV-induced brain injury is unknown, particularly when ultrasound imaging is normal. In a pregnant pigtail macaque (Macaca nemestrina) model of ZIKV infection, we demonstrate that ZIKV-induced injury to fetal brain is substantial, even in the absence of microcephaly, and may be challenging to detect in a clinical setting. A common and subtle injury pattern was identified, including (i) periventricular T2-hyperintense foci and loss of fetal noncortical brain volume, (ii) injury to the ependymal epithelium with underlying gliosis and (iii) loss of late fetal neuronal progenitor cells in the subventricular zone (temporal cortex) and subgranular zone (dentate gyrus, hippocampus) with dysmorphic granule neuron patterning. Attenuation of fetal neurogenic output demonstrates potentially considerable teratogenic effects of congenital ZIKV infection even without microcephaly. Our findings suggest that all children exposed to ZIKV in utero should receive long-term monitoring for neurocognitive deficits, regardless of head size at birth.
- Published
- 2018
- Full Text
- View/download PDF
170. TMOD-13. MODELING THE GENETIC, TRANSCRIPTOMIC, AND CELLULAR HETEROGENEITY OF GLIOBLASTOMA USING TUMOR ORGANOIDS
- Author
-
Fadi Jacob, Guo Li Ming, Ryan D Salinas, Daniel Zhang, Hongjun Song, and Phuong D. Nguyen
- Subjects
Transcriptome ,Cancer Research ,Oncology ,Cellular heterogeneity ,Tumor Models ,medicine ,Neurology (clinical) ,Computational biology ,medicine.disease ,Glioblastoma - Abstract
Glioblastoma exhibits enormous genetic, transcriptional, and cellular heterogeneity at the macroscopic level across regions of the tumor as well as at the microscopic level between neighboring cells, all of which present significant challenges towards creating a definitive treatment for this devastating disease. We have developed a method of generating glioblastoma organoids (GBOs) from fresh tissue obtained directly from surgical resection and maintaining them in a defined medium without bFGF/EGF. Whole exome sequencing revealed that GBOs maintain the genomic landscape of their parent tumors. Somatic and copy number variants are present in the GBOs at similar allele frequencies or copy ratios as in the parent tumor, suggesting that the relative proportions of clonal populations are largely maintained in the organoids. Bulk transcriptomic analysis demonstrated strong gene expression correlations between the parent tumor and corresponding GBOs through 12 weeks of culture. Some tumors were sampled at multiple different anatomic regions, and the corresponding GBOs maintained region-specific gene expression signatures and genomic variants. EGFRvIII, a tumor-specific variant targeted in a number of emerging therapies, also remains present in the GBOs at similar transcript frequencies, reflecting the native heterogeneity of the parent tumor. Finally, we used single cell transcriptomics to examine cellular heterogeneity and find that GBOs contain many different cell types that exhibit similar gene expression profiles as the matching cell type in the corresponding parent tumor. Notably, these GBOs retain neoplastic as well as non-neoplastic cells, such as tumor associated macrophages / microglia, T-cells, endothelial cells, stromal cells, and oligodendrocytes. These GBOs preserve complex tumor heterogeneity an in vitro environment, creating opportunities for extended manipulation, characterization, and functional study for mechanistic investigation and therapeutic testing.
- Published
- 2019
- Full Text
- View/download PDF
171. Interplay between a Mental Disorder Risk Gene and Developmental Polarity Switch of GABA Action Leads to Excitation-Inhibition Imbalance
- Author
-
Benedikt Berninger, Guo Li Ming, Jennifer H. Lee, Jaesuk Park, Qassim Hussani, Shaoyu Ge, Yu-Ting Lin, Hongjun Song, Kuei Sen Hsu, Weidong Li, Juan Song, Yan Gu, Kimberly M. Christian, and Eunchai Kang
- Subjects
Male ,0301 basic medicine ,Neurogenesis ,Mutant ,Nerve Tissue Proteins ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,DISC1 ,Glutamatergic ,0302 clinical medicine ,Risk Factors ,mental disorders ,Animals ,Genetic Predisposition to Disease ,GABAergic Neurons ,gamma-Aminobutyric Acid ,Neurons ,Gene knockdown ,biology ,Mental Disorders ,Cell Polarity ,Neural Inhibition ,Depolarization ,Synaptic Potentials ,Mice, Inbred C57BL ,030104 developmental biology ,nervous system ,Gene Knockdown Techniques ,Synapses ,biology.protein ,Excitatory postsynaptic potential ,GABAergic ,Female ,Neuroscience ,030217 neurology & neurosurgery ,Parvalbumin - Abstract
Excitation-inhibition (E-I) imbalance is considered a hallmark of various neurodevelopmental disorders, including schizophrenia and autism. How genetic risk factors disrupt coordinated glutamatergic and GABAergic synapse formation to cause an E-I imbalance is not well understood. Here, we show that knockdown of Disrupted-in-schizophrenia 1 (DISC1), a risk gene for major mental disorders, leads to E-I imbalance in mature dentate granule neurons. We found that excessive GABAergic inputs from parvalbumin-, but not somatostatin-, expressing interneurons enhance the formation of both glutamatergic and GABAergic synapses in immature mutant neurons. Following the switch in GABAergic signaling polarity from depolarizing to hyperpolarizing during neuronal maturation, heightened inhibition from excessive parvalbumin(+) GABAergic inputs causes loss of excitatory glutamatergic synapses in mature mutant neurons, resulting in an E-I imbalance. Our findings provide insights into the developmental role of depolarizing GABA in establishing E-I balance and how it can be influenced by genetic risk factors for mental disorders.
- Published
- 2019
- Full Text
- View/download PDF
172. FMRP Modulates Neural Differentiation through m6A-Dependent mRNA Nuclear Export
- Author
-
Hongjun Song, Nimrod Miller, Francisca Rojas Ringeling, Guo Li Ming, Saravanan Arumugam, Yong Chao Ma, Caroline Vissers, Zhaofa Xu, Han Shi, Chuan He, Jing Su, and Brittany M. Edens
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Messenger RNA ,Methyltransferase ,RNA ,Biology ,FMR1 ,General Biochemistry, Genetics and Molecular Biology ,Neural stem cell ,nervous system diseases ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Progenitor cell ,Nuclear export signal ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Summary N6-methyladenosine (m6A) modification of mRNA is emerging as a vital mechanism regulating RNA function. Here, we show that fragile X mental retardation protein (FMRP) reads m6A to promote nuclear export of methylated mRNA targets during neural differentiation. Fmr1 knockout (KO) mice show delayed neural progenitor cell cycle progression and extended maintenance of proliferating neural progenitors into postnatal stages, phenocopying methyltransferase Mettl14 conditional KO (cKO) mice that have no m6A modification. RNA-seq and m6A-seq reveal that both Mettl14cKO and Fmr1KO lead to the nuclear retention of m6A-modified FMRP target mRNAs regulating neural differentiation, indicating that both m6A and FMRP are required for the nuclear export of methylated target mRNAs. FMRP preferentially binds m6A-modified RNAs to facilitate their nuclear export through CRM1. The nuclear retention defect can be mitigated by wild-type but not nuclear export-deficient FMRP, establishing a critical role for FMRP in mediating m6A-dependent mRNA nuclear export during neural differentiation.
- Published
- 2019
- Full Text
- View/download PDF
173. Correction to: Rescue of Methyl-CpG Binding Protein 2 Dysfunction-induced Defects in Newborn Neurons by Pentobarbital
- Author
-
Antonius M. J. Van Dongen, Eyleen L. K. Goh, Su In Yoon, Guo Li Ming, Guillaume Marcy, Vinu Ganapathy, Chih Hao Yang, Wan Ying Leong, Dongliang Ma, George J. Augustine, Na Zhao, Ju Han, and Kuei Sen Hsu
- Subjects
Pentobarbital ,medicine.medical_specialty ,Time Factors ,Neurology ,Methyl-CpG-Binding Protein 2 ,Tetrodotoxin ,Pharmacology ,Hippocampus ,Mice ,Text mining ,medicine ,Animals ,Humans ,Rats, Long-Evans ,Pharmacology (medical) ,Cells, Cultured ,gamma-Aminobutyric Acid ,Neurons ,business.industry ,Chemistry ,Correction ,Excitatory Postsynaptic Potentials ,Embryo, Mammalian ,Synapsins ,Rats ,METHYL-CpG-BINDING PROTEIN 2 ,Neurology (clinical) ,Nerve Net ,business ,Adjuvants, Anesthesia ,Signal Transduction ,Sodium Channel Blockers ,medicine.drug - Abstract
Rett syndrome is a neurodevelopmental disorder that usually arises from mutations or deletions in methyl-CpG binding protein 2 (MeCP2), a transcriptional regulator that affects neuronal development and maturation without causing cell loss. Here, we show that silencing of MeCP2 decreased neurite arborization and synaptogenesis in cultured hippocampal neurons from rat fetal brains. These structural defects were associated with alterations in synaptic transmission and neural network activity. Similar retardation of dendritic growth was also observed in MeCP2-deficient newborn granule cells in the dentate gyrus of adult mouse brains in vivo, demonstrating direct and cell-autonomous effects on individual neurons. These defects, caused by MeCP2 deficiency, were reversed by treatment with the US Food and Drug Administration-approved drug, pentobarbital, in vitro and in vivo, possibly caused by modulation of γ-aminobutyric acid signaling. The results indicate that drugs modulating γ-aminobutyric acid signaling are potential therapeutics for Rett syndrome.
- Published
- 2019
- Full Text
- View/download PDF
174. A Common Embryonic Origin of Stem Cells Drives Developmental and Adult Neurogenesis
- Author
-
Dennisse Jimenez-Cyrus, Jonathan A. Epstein, Hongjun Song, Guo Li Ming, David Morizet, Stephanie Lee, Rajan Jain, Stefan Canzar, Francisca Rojas Ringeling, Daniel A. Berg, Nancy Huang, Reeti Shah, Qing-Feng Wu, Yijing Su, Allison M. Bond, and Aneek Patel
- Subjects
Male ,Neurogenesis ,Population ,Mice, Transgenic ,Biology ,Hippocampus ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,Animals ,education ,Embryonic Stem Cells ,030304 developmental biology ,Progenitor ,Homeodomain Proteins ,0303 health sciences ,education.field_of_study ,Dentate gyrus ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Embryo, Mammalian ,Embryonic stem cell ,Neural stem cell ,Mice, Inbred C57BL ,Neuroepithelial cell ,Dentate Gyrus ,Female ,Stem cell ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Summary New neurons arise from quiescent adult neural progenitors throughout life in specific regions of the mammalian brain. Little is known about the embryonic origin and establishment of adult neural progenitors. Here, we show that Hopx+ precursors in the mouse dentate neuroepithelium at embryonic day 11.5 give rise to proliferative Hopx+ neural progenitors in the primitive dentate region, and they, in turn, generate granule neurons, but not other neurons, throughout development and then transition into Hopx+ quiescent radial glial-like neural progenitors during an early postnatal period. RNA-seq and ATAC-seq analyses of Hopx+ embryonic, early postnatal, and adult dentate neural progenitors further reveal common molecular and epigenetic signatures and developmental dynamics. Together, our findings support a "continuous" model wherein a common neural progenitor population exclusively contributes to dentate neurogenesis throughout development and adulthood. Adult dentate neurogenesis may therefore represent a lifelong extension of development that maintains heightened plasticity in the mammalian hippocampus.
- Published
- 2019
- Full Text
- View/download PDF
175. Persistent Cyfipl Expression Is Required to Maintain the Adult Subventricular Zone Neurogenic Niche.
- Author
-
Habela, Christa Whelan, Ki-Jun Yoon, Nam-Shik Kim, Taga, Arens, Bell, Kassidy, Bergles, Dwight E., Maragakis, Nicholas J., Guo-li Ming, and Hongjun Song
- Subjects
NEURAL stem cells ,LIFE zones ,ADHERENS junctions ,CELL proliferation ,EMBRYOLOGY - Abstract
Neural stem cells (NSCs) persist throughout life in the subventricular zone (SVZ) neurogenic niche of the lateral ventricles as Type B1 cells in adult mice. Maintaining this population of NSCs depends on the balance between quiescence and self-renewing or self-depleting cell divisions. Interactions between B1 cells and the surrounding niche are important in regulating this balance, but the mechanisms governing these processes have not been fully elucidated. The cytoplasmic FMRP-interacting protein (Cyfip1) regulates apical-basal polarity in the embryonic brain. Loss of Cyfip1 during embryonic development in mice disrupts the embryonic niche and affects cortical neurogenesis. However, a direct role for Cyfip1 in the regulation of adult NSCs has not been established. Here, we demonstrate that Cyfip1 expression is preferentially localized to B1 cells in the adult mouse SVZ. Loss of Cyfip1 in the embryonic mouse brain results in altered adult SVZ architecture and expansion of the adult B1 cell population at the ventricular surface. Furthermore, acute deletion of Cyfip1 in adult NSCs results in a rapid change in adherens junction proteins as well as increased proliferation and number of B1 cells at the ventricular surface. Together, these data indicate that Cyfip1 plays a critical role in the formation and maintenance of the adult SVZ niche; furthermore, deletion of Cyfip1 unleashes the capacity of adult B1 cells for symmetric renewal to increase the adult NSC pool.SIGNIFICANCE STATEMENT Neural stem cells (NSCs) persist in the subventricular zone of the lateral ventricles in adult mammals, and the size of this population is determined by the balance between quiescence and self-depleting or renewing cell division. The mechanisms regulating these processes are not fully understood. This study establishes that the cytoplasmic FMRP interacting protein 1 (Cyfip1) regulates NSC fate decisions in the adult subventricular zone and adult NSCs that are quiescent or typically undergo self-depleting divisions retain the ability to self-renew. These results contribute to our understanding of how adult NSCs are regulated throughout life and has potential implications for human brain disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
176. Latent tri-lineage potential of adult hippocampal neural stem cells revealed by Nf1 inactivation
- Author
-
Genevieve Stein-O’Brien, Michael A. Bonaguidi, Shiori Ito, Yi Zhou, Nicholas K. Kawasaki, Guo Li Ming, Hongjun Song, Yuan Zhu, Nikhil Modak, and Gerald J. Sun
- Subjects
Lineage (genetic) ,Cellular differentiation ,Mice, Transgenic ,Hippocampus ,Article ,Mice ,medicine ,Animals ,Cell Lineage ,Neurofibromin 1 ,biology ,General Neuroscience ,Neurogenesis ,Cell Differentiation ,Oligodendrocyte ,Neural stem cell ,nervous system diseases ,Mice, Inbred C57BL ,Adult Stem Cells ,Oligodendroglia ,medicine.anatomical_structure ,nervous system ,biology.protein ,Stem cell ,Neuroscience ,Adult stem cell - Abstract
Endogenous neural stem cells (NSCs) in the adult hippocampus are considered to be bi-potent, as they only produce neurons and astrocytes in vivo. In mouse, we found that inactivation of neurofibromin 1 (Nf1), a gene mutated in neurofibromatosis type 1, unlocked a latent oligodendrocyte lineage potential to produce all three lineages from NSCs in vivo. Our results suggest an avenue for promoting stem cell plasticity by targeting barriers of latent lineage potential.
- Published
- 2015
177. DNA Modifications and Neurological Disorders
- Author
-
Ran An, Guo Li Ming, Jaehoon Shin, Yi Lan Weng, and Hongjun Song
- Subjects
Neurons ,Pharmacology ,Regulation of gene expression ,Genetics ,Epigenetics in learning and memory ,Brain ,Review ,DNA Methylation ,Biology ,Epigenesis, Genetic ,Cytosine ,Epigenetics of physical exercise ,Gene Expression Regulation ,DNA methylation ,5-Methylcytosine ,Humans ,Pharmacology (medical) ,Neurology (clinical) ,Cancer epigenetics ,Epigenetics ,Nervous System Diseases ,RNA-Directed DNA Methylation ,Epigenomics - Abstract
Mounting evidence has recently underscored the importance of DNA methylation in normal brain functions. DNA methylation machineries are responsible for dynamic regulation of methylation patterns in discrete brain regions. In addition to methylation of cytosines in genomic DNA (5-methylcytosine; 5mC), other forms of modified cytosines, such as 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine, can potentially act as epigenetic marks that regulate gene expression. Importantly, epigenetic modifications require cognate binding proteins to read and translate information into gene expression regulation. Abnormal or incorrect interpretation of DNA methylation patterns can cause devastating consequences, including mental illnesses and neurological disorders. Although DNA methylation was generally considered to be a stable epigenetic mark in post-mitotic cells, recent studies have revealed dynamic DNA modifications in neurons. Such reversibility of 5mC sheds light on potential mechanisms underlying some neurological disorders and suggests a new route to correct aberrant methylation patterns associated with these disorders.
- Published
- 2013
- Full Text
- View/download PDF
178. TET1 Controls CNS 5-Methylcytosine Hydroxylation, Active DNA Demethylation, Gene Transcription, and Memory Formation
- Author
-
Dawn E. Eason, J. David Sweatt, Daniel L. Ross, Jennifer R. King, Hongjun Song, Guo Li Ming, Raj V. Vachhani, Garrett A. Kaas, and Chun Zhong
- Subjects
Central Nervous System ,Male ,Time Factors ,Transcription, Genetic ,Epigenetics in learning and memory ,Neuroscience(all) ,Conditioning, Classical ,Mutant ,Convulsants ,Nerve Tissue Proteins ,Motor Activity ,Biology ,Hydroxylation ,Hippocampus ,Chrysenes ,Article ,Adenoviridae ,Mixed Function Oxygenases ,Potassium Chloride ,Cytosine ,Mice ,chemistry.chemical_compound ,Downregulation and upregulation ,Memory ,Seizures ,Transduction, Genetic ,Proto-Oncogene Proteins ,Flurothyl ,Animals ,Gene ,Cells, Cultured ,Neurons ,Regulation of gene expression ,Analysis of Variance ,General Neuroscience ,DNA Methylation ,Molecular biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Luminescent Proteins ,5-Methylcytosine ,DNA demethylation ,Animals, Newborn ,Gene Expression Regulation ,chemistry ,Mutation ,DNA methylation - Abstract
Summary Dynamic changes in 5-methylcytosine (5mC) have been implicated in the regulation of gene expression critical for consolidation of memory. However, little is known about how these changes in 5mC are regulated in the adult brain. The enzyme methylcytosine dioxygenase TET1 (TET1) has been shown to promote active DNA demethylation in the nervous system. Therefore, we took a viral-mediated approach to overexpress the protein in the hippocampus and examine its potential involvement in memory formation. We found that Tet1 is a neuronal activity-regulated gene and that its overexpression leads to global changes in modified cytosine levels. Furthermore, expression of TET1 or a catalytically inactive mutant (TET1m) resulted in the upregulation of several neuronal memory-associated genes and impaired contextual fear memory. In summary, we show that neuronal Tet1 regulates DNA methylation levels and that its expression, independent of its catalytic activity, regulates the expression of CNS activity-dependent genes and memory formation.
- Published
- 2013
- Full Text
- View/download PDF
179. Seamless Reconstruction of Intact Adult-Born Neurons by Serial End-Block Imaging Reveals Complex Axonal Guidance and Development in the Adult Hippocampus
- Author
-
Nikhil Chavali, Hongjun Song, Gerald J. Sun, Kurt A. Sailor, Kimberly M. Christian, Qasim A. Mahmood, and Guo Li Ming
- Subjects
Neurons ,Mouse Hippocampus ,Nervous system ,Microscopy, Confocal ,Neurogenesis ,General Neuroscience ,Age Factors ,Articles ,Hippocampal formation ,Biology ,Hippocampus ,Axons ,Mice, Inbred C57BL ,Mice ,medicine.anatomical_structure ,Animals, Newborn ,nervous system ,Initial phase ,medicine ,Animals ,Female ,Neuron ,Neuroscience ,Tissue volume - Abstract
In the adult mammalian hippocampus, newborn dentate granule cells are continuously integrated into the existing circuitry and contribute to specific brain functions. Little is known about the axonal development of these newborn neurons in the adult brain due to technological challenges that have prohibited large-scale reconstruction of long, thin, and complex axonal processes within the mature nervous system. Here, using a new serial end-block imaging (SEBI) technique, we seamlessly reconstructed axonal and dendritic processes of intact individual retrovirus-labeled newborn granule cells at different developmental stages in the young adult mouse hippocampus. We found that adult-born dentate granule cells exhibit tortuous, yet highly stereotyped, axonal projections to CA3 hippocampal subregions. Primary axonal projections of cohorts of new neurons born around the same time organize into laminar patterns with staggered terminations that stack along the septo-temporal hippocampal axis. Analysis of individual newborn neuron development further defined an initial phase of rapid axonal and dendritic growth within 21 d after newborn neuron birth, followed by minimal growth of primary axonal and whole dendritic processes through the last time point examined at 77 d. Our results suggest that axonal development and targeting is a highly orchestrated, precise process in the adult brain. These findings demonstrate a striking regenerative capacity of the mature CNS to support long-distance growth and guidance of neuronal axons. Our SEBI approach can be broadly applied for analysis of intact, complex neuronal projections in limitless tissue volume.
- Published
- 2013
- Full Text
- View/download PDF
180. DISC1 and SLC12A2 interaction affects human hippocampal function and connectivity
- Author
-
Karen F. Berman, Michael G. White, Joseph H. Callicott, David A.A. Baranger, Guo Li Ming, Qiang Chen, Bai Lu, Emer L. Feighery, Hongjun Song, Daniel R. Weinberger, and Venkata S. Mattay
- Subjects
Adult ,Heterozygote ,Adolescent ,Sodium-Potassium-Chloride Symporters ,Hippocampus ,Nerve Tissue Proteins ,Hippocampal formation ,Neurotransmission ,Polymorphism, Single Nucleotide ,Young Adult ,DISC1 ,Connectome ,medicine ,Humans ,Solute Carrier Family 12, Member 2 ,Genetic Predisposition to Disease ,Recognition memory ,Genetics ,biology ,Brief Report ,Epistasis, Genetic ,Depolarization ,General Medicine ,Middle Aged ,Phenotype ,medicine.anatomical_structure ,Schizophrenia ,biology.protein ,Parahippocampal Gyrus ,Neuroscience ,Parahippocampal gyrus - Abstract
Hippocampal development is coordinated by both extracellular factors like GABA neurotransmission and intracellular components like DISC1. We previously reported that SLC12A2-dependent GABA depolarization and DISC1 coregulate hippocampal neuronal development, and 2 SNPs in these genes linked to mRNA expression interactively increase schizophrenia risk. Using functional MRI, we now confirm this biological interaction in vivo by showing in 2 independent samples of healthy individuals (total N = 349) that subjects homozygous for both risk alleles evince dramatically decreased hippocampal area activation (Cohen’s d = 0.78) and connectivity (d = 0.57) during a recognition memory task. These data highlight the importance of epistatic models in understanding genetic association with complex brain phenotypes.
- Published
- 2013
- Full Text
- View/download PDF
181. Secreted Frizzled-Related Protein 3 Regulates Activity-Dependent Adult Hippocampal Neurogenesis
- Author
-
Yijing Su, Marie Xun Wang, Yuan Gao, Ju Young Kim, Kurt A. Sailor, Michael A. Bonaguidi, Guo Li Ming, Hongjun Song, Juan Song, Jiaqi Sun, Heechul Jun, Kimberly M. Christian, Chun Zhong, Yasuji Kitabatake, Junjie U. Guo, Mi Hyeon Jang, and Eunchai Kang
- Subjects
Dendritic spine ,Dentate gyrus ,Neurogenesis ,Cell Biology ,Anatomy ,Hippocampal formation ,Biology ,Neural stem cell ,Cell biology ,medicine.anatomical_structure ,Neuropoiesis ,nervous system ,Genetics ,Neuron maturation ,medicine ,Molecular Medicine ,Neuron - Abstract
SummaryAdult neurogenesis, the process of generating mature neurons from adult neural stem cells, proceeds concurrently with ongoing neuronal circuit activity and is modulated by various physiological and pathological stimuli. The niche mechanism underlying the activity-dependent regulation of the sequential steps of adult neurogenesis remains largely unknown. Here, we report that neuronal activity decreases the expression of secreted frizzled-related protein 3 (sFRP3), a naturally secreted Wnt inhibitor highly expressed by adult dentate gyrus granule neurons. Sfrp3 deletion activates quiescent radial neural stem cells and promotes newborn neuron maturation, dendritic growth, and dendritic spine formation in the adult mouse hippocampus. Furthermore, sfrp3 reduction is essential for activity-induced adult neural progenitor proliferation and the acceleration of new neuron development. Our study identifies sFRP3 as an inhibitory niche factor from local mature dentate granule neurons that regulates multiple phases of adult hippocampal neurogenesis and suggests an interesting activity-dependent mechanism governing adult neurogenesis via the acute release of tonic inhibition.
- Published
- 2013
- Full Text
- View/download PDF
182. Enhancing oligodendrocyte differentiation by transient transcription activation via DNA nanoparticle-mediated transfection
- Author
-
Guo Li Ming, Xiaowei Li, Vassilis E. Koliatsos, Hai-Quan Mao, Jordan J. Green, Camila Gadens Zamboni, and Stephany Y. Tzeng
- Subjects
0301 basic medicine ,Pluripotent Stem Cells ,Transcriptional Activation ,Biomedical Engineering ,Nerve Tissue Proteins ,Biology ,Transfection ,Biochemistry ,Article ,Biomaterials ,OLIG2 ,03 medical and health sciences ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Humans ,Remyelination ,Progenitor cell ,Induced pluripotent stem cell ,Molecular Biology ,Oligodendrocyte differentiation ,Cell Differentiation ,General Medicine ,Oligodendrocyte Transcription Factor 2 ,Molecular biology ,Oligodendrocyte ,Cell biology ,Oligodendroglia ,030104 developmental biology ,medicine.anatomical_structure ,Nanoparticles ,Stem cell ,Biotechnology - Abstract
Current approaches to derive oligodendrocytes from human pluripotent stem cells (hPSCs) need extended exposure of hPSCs to growth factors and small molecules, which limits their clinical application because of the lengthy culture time required and low generation efficiency of myelinating oligodendrocytes. Compared to extrinsic growth factors and molecules, oligodendrocyte differentiation and maturation can be more effectively modulated by regulation of the cell transcription network. In the developing central nervous system (CNS), two basic helix-loop-helix transcription factors, Olig1 and Olig2, are decisive in oligodendrocyte differentiation and maturation. Olig2 plays a critical role in the specification of oligodendrocytes and Olig1 is crucial in promoting oligodendrocyte maturation. Recently viral vectors have been used to overexpress Olig2 and Olig1 in neural stem/progenitor cells (NSCs) to induce the maturation of oligodendrocytes and enhance the remyelination activity in vivo . Because of the safety issues with viral vectors, including the insertional mutagenesis and potential tumor formation, non-viral transfection methods are preferred for clinical translation. Here we report a poly(β-amino ester) (PBAE)-based nanoparticle transfection method to deliver Olig1 and Olig2 into human fetal tissue-derived NSCs and demonstrate efficient oligodendrocyte differentiation following transgene expression of Olig1 and Olig2. This approach is potentially translatable for engineering stem cells to treat injured or diseased CNS tissues. Statement of Significance Current protocols to derive oligodendrocytes from human pluripotent stem cells (hPSCs) require lengthy culture time with low generation efficiencies of mature oligodendrocytes. We described a new approach to enhance oligodendrocyte differentiation through nanoparticle-mediated transcription modulation. We tested an effective transfection method using cell-compatible poly (β-amino ester) (PBAE)/DNA nanoparticles as gene carrier to deliver transcription factor Olig1 and Olig2 into human fetal tissue-derived neural stem/progenitor cells, and showed efficient oligodendrocyte differentiation following transgene expression of Olig1 and Olig2. We believe that this translatable approach can be applied to many other cell-based regenerative therapies as well.
- Published
- 2016
183. A nuclease that mediates cell death induced by DNA damage and poly(ADP-ribose) polymerase-1
- Author
-
Jun Seop Jeong, Stewart Neifert, Jiang Qian, Ted M. Dawson, Bong Woo Kim, Kalyani Nambiar, Guo Li Ming, Yingfei Wang, Lei Bao, Maged M. Harraz, Hyejin Park, Hongjun Song, Shaida A. Andrabi, Zhi Xie, Heng Zhu, Tae In Kam, Calvin Chang, Jennifer E. Wang, Seth Blackshaw, Valina L. Dawson, Rong Chen, Ran An, Stephen M. Eacker, and George K.E. Umanah
- Subjects
0301 basic medicine ,Programmed cell death ,DNA damage ,Poly ADP ribose polymerase ,Active Transport, Cell Nucleus ,Poly (ADP-Ribose) Polymerase-1 ,DNA, Single-Stranded ,Glutamic Acid ,Apoptosis ,DNA Fragmentation ,Biology ,Mice ,03 medical and health sciences ,Endonuclease ,Catalytic Domain ,Animals ,Humans ,Amino Acid Sequence ,DNA Cleavage ,Macrophage Migration-Inhibitory Factors ,Cell Nucleus ,Mice, Knockout ,Neurons ,Deoxyribonucleases ,Multidisciplinary ,Base Sequence ,Apoptosis Inducing Factor ,Molecular biology ,Chromatin ,Mitochondria ,Intramolecular Oxidoreductases ,Stroke ,Oxidative Stress ,030104 developmental biology ,Mutation ,biology.protein ,Nucleic Acid Conformation ,DNA fragmentation ,Macrophage migration inhibitory factor ,DNA Damage ,HeLa Cells - Abstract
DNA damage-activated nuclease identified Cells that experience stresses and accumulate excessive damage to DNA undergo cell death mediated by a nuclear enzyme known as PARP-1. During this process, apoptosis-inducing factor (AIF) translocates to the nucleus and activates one or more nucleases to cleave DNA. Wang et al. found that macrophage migration inhibitory factor (MIF) is an AIF-associated endonuclease that contributes to PARP-1-induced DNA fragmentation (see the Perspective by Jonas). In mouse neurons in culture, loss of MIF protected neurons from cell death caused by excessive stimulation. Targeting MIF could thus provide a therapeutic strategy against diseases in which PARP-1 activation is excessive. Science , this issue p. 82 ; see also p. 36
- Published
- 2016
- Full Text
- View/download PDF
184. Early postnatal exposure to isoflurane causes cognitive deficits and disrupts development of newborn hippocampal neurons via activation of the mTOR pathway
- Author
-
Yun Kyoung Ryu, Jieun Kim, Minhye Kwak, Jun H. Choi, Danye Jiang, Sanghee Lim, Sue Junn, Roger A. Johns, Guo Li Ming, Hongjun Song, Jing Xu, Eunchai Kang, Christy D. Gray, Michele L. Schaefer, C. David Mintz, and Michael Xu
- Subjects
Dendritic spine ,QH301-705.5 ,Dendritic Spines ,Hippocampal formation ,Biology ,Hippocampus ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,030202 anesthesiology ,medicine ,Animals ,Cognitive Dysfunction ,Biology (General) ,PI3K/AKT/mTOR pathway ,Neurons ,General Immunology and Microbiology ,Isoflurane ,General Neuroscience ,Dentate gyrus ,TOR Serine-Threonine Kinases ,Correction ,Environmental exposure ,Environmental Exposure ,Granule cell ,3. Good health ,medicine.anatomical_structure ,Anesthetic ,Anesthetics, Inhalation ,General Agricultural and Biological Sciences ,Neuroscience ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Clinical and preclinical studies indicate that early postnatal exposure to anesthetics can lead to lasting deficits in learning and other cognitive processes. The mechanism underlying this phenomenon has not been clarified and there is no treatment currently available. Recent evidence suggests that anesthetics might cause persistent deficits in cognitive function by disrupting key events in brain development. The hippocampus, a brain region that is critical for learning and memory, contains a large number of neurons that develop in the early postnatal period, which are thus vulnerable to perturbation by anesthetic exposure. Using an in vivo mouse model we demonstrate abnormal development of dendrite arbors and dendritic spines in newly generated dentate gyrus granule cell neurons of the hippocampus after a clinically relevant isoflurane anesthesia exposure conducted at an early postnatal age. Furthermore, we find that isoflurane causes a sustained increase in activity in the mechanistic target of rapamycin pathway, and that inhibition of this pathway with rapamycin not only reverses the observed changes in neuronal development, but also substantially improves performance on behavioral tasks of spatial learning and memory that are impaired by isoflurane exposure. We conclude that isoflurane disrupts the development of hippocampal neurons generated in the early postnatal period by activating a well-defined neurodevelopmental disease pathway and that this phenotype can be reversed by pharmacologic inhibition.
- Published
- 2016
185. Molecular signatures associated with ZIKV exposure in human cortical neural progenitors
- Author
-
Zhiqin Wang, Ha Nam Nguyen, Yichen Cheng, Li Chen, Miao Xu, Hengli Tang, Xuyu Qian, Tianlei Xu, Zhexing Wen, Pei Yong Shi, Guo Li Ming, Emily M. Lee, Menghang Xia, Ki Jun Yoon, Kimberly M. Christian, Hao Wu, Wei Zheng, Christy Hammack, Mingjiang Xu, Luoxiu Huang, Feiran Zhang, Sarah C. Ogden, Wei Kai Huang, Chao Shan, Bing Yao, Hongjun Song, Yujing Li, Zhaohui S. Qin, Hao Feng, and Peng Jin
- Subjects
DNA Replication ,0301 basic medicine ,Zika virus disease ,Microcephaly ,DNA Repair ,viruses ,Data Resources and Analyses ,Biology ,Dengue virus ,medicine.disease_cause ,Virus ,Cell Line ,Zika virus ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,Species Specificity ,Genetics ,medicine ,Humans ,Progenitor cell ,Gene ,030304 developmental biology ,Cerebral Cortex ,0303 health sciences ,Cell Death ,Zika Virus Infection ,Gene Expression Profiling ,Zika Virus ,Dengue Virus ,biology.organism_classification ,medicine.disease ,Virology ,Neural stem cell ,Up-Regulation ,Gene expression profiling ,030104 developmental biology ,Tumor Suppressor Protein p53 ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Zika virus (ZIKV) infection causes microcephaly and has been linked to other brain abnormalities. How ZIKV impairs brain development and function is unclear. Here we systematically profiled transcriptomes of human neural progenitor cells exposed to Asian ZIKVC, African ZIKVM, and dengue virus (DENV). In contrast to the robust global transcriptome changes induced by DENV, ZIKV has a more selective and larger impact on expression of genes involved in DNA replication and repair. While overall expression profiles are similar, ZIKVC, but not ZIKVM, induces upregulation of viral response genes and TP53. P53 inhibitors can block the apoptosis induced by both ZIKVC and ZIKVM in hNPCs, with higher potency against ZIKVC-induced apoptosis. Our analyses reveal virus- and strain-specific molecular signatures associated with ZIKV infection. These datasets will help to investigate ZIKV-host interactions and identify neurovirulence determinants of ZIKV.
- Published
- 2016
- Full Text
- View/download PDF
186. Persistent Structural Plasticity Optimizes Sensory Information Processing in the Olfactory Bulb
- Author
-
Matthew T. Valley, Guo Li Ming, Gerald J. Sun, Hermann Riecke, Shirin Sharafi, Pierre-Marie Lledo, Martin T. Wiechert, Wayne Adams, Kurt A. Sailor, James C. Dennis, Hongjun Song, Perception et Mémoire / Perception and Memory, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Institute for Cell Engineering [Baltimore] (ICE), Johns Hopkins University (JHU)-Johns Hopkins University School of Medicine [Baltimore], Diana Helis Henry Medical Research Foundation [New Orleans], The Solomon H. Snyder Department of Neuroscience [Baltimore], Department of Engineering Sciences and Applied Mathematics [Evanston, USA], McCormick School of Engineering and Applied Mathematics [Evanston, USA], Northwestern University [Evanston]-Northwestern University [Evanston], Johns Hopkins University School of Medicine [Baltimore], Support was provided by the following: W.A. and J.C.D., NSF undergraduate support RTG DMS-0636574, K.A.S., P.M.L., M.T.V., and M.T.W., 'AG2R-La-Mondiale' life insurance, Agence Nationale de la Recherche ANR-15-CE37-0004-01, NIH US-French Research Proposal Grants #1R01DC015137-01 and ANR-15-NEUC-0004 (Circuit-OPL), the Laboratory for Excellence 'Revive' Program (Investissement d’Avenir, ANR-10-LABX-73) and 'Biopsy' (Investissement d’Avenir, ANR-11-IDEX-0004-02), K.A.S., NIH NRSA F31NS066612 and 'Revive' fellowships, M.T.V., Pasteur-Roux fellowship and the Phillippe Foundation. H.J.S., NIH R37NS047344, G.L.M., NIH R01MH105128 and NIH R01NS048271, and the authors acknowledge the joint participation by the Diana Helis Henry Medical Research Foundation through its direct engagement in the continuous active conduct of medical research in conjunction with The Johns Hopkins Hospital and the Johns Hopkins University School of Medicine and the Foundation’s Parkinson’s Disease Program (No. H-1)., We thank Eileen Huang for data analysis, Soham Saha, Camille Mazo, Gabriel Lepousez, and Cyrille Norotte for manual puncta analysis, Dwight Bergels, David Linden, Randall Reed, Richard Huganir, and Chun Zhong for advice, Anne Lanjuin for providing Tbet-Cre mice, Elly Nedivi and Jerry Chen for providing the gephyrin-teal construct, and David DiGregorio for managing the Pasteur Institute Shared Neuroscience Department imaging facility funded by 'Ile de France Domaine d’Intérêt Majeur (NeRF).' HPC Northwestern University provided HPC access (Quest)., ANR-15-NEUC-0004,CIRCUIT-OPL,US-French Research Proposal- Neural Circuits and Plasticity- Olfactory Perception and Learning(2015), ANR-10-LABX-0073,REVIVE,Stem Cells in Regenerative Biology and Medicine(2010), ANR-11-IDEX-0004,SUPER,Sorbonne Universités à Paris pour l'Enseignement et la Recherche(2011), and Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Interneurons / physiology ,Patch-Clamp Techniques ,Interneuron ,Dendritic Spines ,Neurogenesis ,Nonsynaptic plasticity ,Article ,03 medical and health sciences ,Neuronal Plasticity / physiology ,Mice ,0302 clinical medicine ,Interneurons ,Metaplasticity ,medicine ,Animals ,Synaptic scaling ,Neuronal Plasticity ,Homosynaptic plasticity ,Neurogenesis / physiology ,General Neuroscience ,[SCCO.NEUR]Cognitive science/Neuroscience ,Nerve Net / metabolism ,Synapses / metabolism ,Dendritic Spines / metabolism ,Olfactory Bulb ,030104 developmental biology ,Synaptic fatigue ,medicine.anatomical_structure ,Olfactory Bulb / physiology ,Synaptic plasticity ,Synapses ,Developmental plasticity ,Nerve Net ,Psychology ,Neuroscience ,030217 neurology & neurosurgery - Abstract
International audience; In the mammalian brain, the anatomical structure of neural circuits changes little during adulthood. As a result, adult learning and memory are thought to result from specific changes in synaptic strength. A possible exception is the olfactory bulb (OB), where activity guides interneuron turnover throughout adulthood. These adult-born granule cell (GC) interneurons form new GABAergic synapses that have little synaptic strength plasticity. In the face of persistent neuronal and synaptic turnover, how does the OB balance flexibility, as is required for adapting to changing sensory environments, with perceptual stability? Here we show that high dendritic spine turnover is a universal feature of GCs, regardless of their developmental origin and age. We find matching dynamics among postsynaptic sites on the principal neurons receiving the new synaptic inputs. We further demonstrate in silico that this coordinated structural plasticity is consistent with stable, yet flexible, decorrelated sensory representations. Together, our study reveals that persistent, coordinated synaptic structural plasticity between interneurons and principal neurons is a major mode of functional plasticity in the OB.
- Published
- 2016
- Full Text
- View/download PDF
187. Epigenetic mechanisms in neurogenesis
- Author
-
Kimberly M. Christian, Guo Li Ming, Chuan He, Bing Yao, Hongjun Song, and Peng Jin
- Subjects
0301 basic medicine ,Neurons ,Epigenetic regulation of neurogenesis ,biology ,General Neuroscience ,Neurogenesis ,Brain ,Embryonic stem cell ,Neural stem cell ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,030104 developmental biology ,Histone ,Neural Stem Cells ,Epitranscriptomics ,biology.protein ,Animals ,Humans ,Epigenetics ,Neuroscience ,Neuroglia ,Epigenesis - Abstract
In the embryonic and adult brain, neural stem cells proliferate and give rise to neurons and glia through highly regulated processes. Epigenetic mechanisms - including DNA and histone modifications, as well as regulation by non-coding RNAs - have pivotal roles in different stages of neurogenesis. Aberrant epigenetic regulation also contributes to the pathogenesis of various brain disorders. Here, we review recent advances in our understanding of epigenetic regulation in neurogenesis and its dysregulation in brain disorders, including discussion of newly identified DNA cytosine modifications. We also briefly cover the emerging field of epitranscriptomics, which involves modifications of mRNAs and long non-coding RNAs.
- Published
- 2016
188. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen
- Author
-
Hengli Tang, Jennifer Kouznetsova, Paul Shinn, Wei Zheng, Wei Kai Huang, Menghang Xia, Miao Xu, Christy Hammack, Julia Tcw, Chase Allen, Xuyu Qian, Alison Goate, Ha Nam Nguyen, Zhexing Wen, Wenwei Huang, Guo Li Ming, Hongjun Song, Samuel G. Michael, Kristen J. Brennand, Sarah C. Ogden, Ruili Huang, Anton Simeonov, Yichen Cheng, Catherine Hanna, Kimberly M. Christian, Fadi Jacob, Misha Itkin, and Emily M. Lee
- Subjects
0301 basic medicine ,Drug ,Programmed cell death ,media_common.quotation_subject ,Induced Pluripotent Stem Cells ,Pharmacology ,Virus Replication ,General Biochemistry, Genetics and Molecular Biology ,Article ,Zika virus ,Cell Line ,03 medical and health sciences ,Neural Stem Cells ,medicine ,Humans ,Pentanoic Acids ,Niclosamide ,media_common ,Neurons ,biology ,Cell Death ,Kinase ,Caspase 3 ,Zika Virus Infection ,Drug Repositioning ,Brain ,General Medicine ,Zika Virus ,biology.organism_classification ,Virology ,Caspase Inhibitors ,Organoids ,Drug repositioning ,030104 developmental biology ,Drug development ,Cell culture ,Astrocytes ,Microcephaly ,medicine.drug - Abstract
In response to the current global health emergency posed by the Zika virus (ZIKV) outbreak and its link to microcephaly and other neurological conditions, we performed a drug repurposing screen of ∼6,000 compounds that included approved drugs, clinical trial drug candidates and pharmacologically active compounds; we identified compounds that either inhibit ZIKV infection or suppress infection-induced caspase-3 activity in different neural cells. A pan-caspase inhibitor, emricasan, inhibited ZIKV-induced increases in caspase-3 activity and protected human cortical neural progenitors in both monolayer and three-dimensional organoid cultures. Ten structurally unrelated inhibitors of cyclin-dependent kinases inhibited ZIKV replication. Niclosamide, a category B anthelmintic drug approved by the US Food and Drug Administration, also inhibited ZIKV replication. Finally, combination treatments using one compound from each category (neuroprotective and antiviral) further increased protection of human neural progenitors and astrocytes from ZIKV-induced cell death. Our results demonstrate the efficacy of this screening strategy and identify lead compounds for anti-ZIKV drug development.
- Published
- 2016
189. Brain Region-specific Organoids using Mini-bioreactors for Modeling ZIKV Exposure
- Author
-
Yujing Li, Hao Wu, Guo Li Ming, William J. Jeang, Michael Chickering, Li Lin, Sarah C. Ogden, Chun Zhong, Ce Zhang, Gregory R. Hamersky, Bing Yao, Xuyu Qian, Jai Thakor, Zhexing Wen, Cheng-Ying Ho, Hengli Tang, Pei Yong Shi, Brady J. Maher, Fadi Jacob, Christopher Hadiono, Ki Jun Yoon, Peng Jin, Mingxi M. Song, Ha Nam Nguyen, Kimberly M. Christian, Eunchai Kang, Christy Hammack, Hongjun Song, Daniel A. Berg, and David W. Nauen
- Subjects
0301 basic medicine ,Zika Virus Infection ,Neurogenesis ,Brain ,Human brain ,Zika Virus ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell biology ,Organoids ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Neural Stem Cells ,Forebrain ,Immunology ,Organoid ,medicine ,Humans ,Progenitor cell ,Induced pluripotent stem cell ,Progenitor ,Cerebral organoid - Abstract
Cerebral organoids, three-dimensional cultures that model organogenesis, provide a new platform to investigate human brain development. High cost, variability, and tissue heterogeneity limit their broad applications. Here, we developed a miniaturized spinning bioreactor (SpinΩ) to generate forebrain-specific organoids from human iPSCs. These organoids recapitulate key features of human cortical development, including progenitor zone organization, neurogenesis, gene expression, and, notably, a distinct human-specific outer radial glia cell layer. We also developed protocols for midbrain and hypothalamic organoids. Finally, we employed the forebrain organoid platform to model Zika virus (ZIKV) exposure. Quantitative analyses revealed preferential, productive infection of neural progenitors with either African or Asian ZIKV strains. ZIKV infection leads to increased cell death and reduced proliferation, resulting in decreased neuronal cell-layer volume resembling microcephaly. Together, our brain-region-specific organoids and SpinΩ provide an accessible and versatile platform for modeling human brain development and disease and for compound testing, including potential ZIKV antiviral drugs.
- Published
- 2016
190. Heterogeneity of Radial Glia-Like Cells in the Adult Hippocampus
- Author
-
Sébastien Sultan, Guo Li Ming, Ruth Beckervordersandforth, Nicolas Toni, Elias Gebara, Florian Udry, Hongjun Song, Pieter Jan Gijs, Dieter Chichung Lie, and Michael A. Bonaguidi
- Subjects
0301 basic medicine ,Neurogenesis ,Cellular differentiation ,Ependymoglial Cells ,Biology ,Animals ,Biomarkers/metabolism ,Cell Lineage/genetics ,Cell Proliferation ,Ependymoglial Cells/cytology ,Ependymoglial Cells/metabolism ,Ependymoglial Cells/transplantation ,Hippocampus/cytology ,Hippocampus/pathology ,Humans ,Mice ,Neural Stem Cells/cytology ,Neural Stem Cells/metabolism ,Neural Stem Cells/transplantation ,Adult stem cells ,Nervous system ,Neural stem cell ,Somatic stem cells ,Stem cell-microenvironment interactions ,Stem cell marker ,Hippocampus ,Article ,03 medical and health sciences ,Neural Stem Cells ,Neurosphere ,Cell Lineage ,Cell Biology ,Anatomy ,Cell biology ,Neuroepithelial cell ,030104 developmental biology ,Molecular Medicine ,Stem cell ,Biomarkers ,Developmental Biology ,Adult stem cell - Abstract
Adult neurogenesis is tightly regulated by the neurogenic niche. Cellular contacts between niche cells and neural stem cells are hypothesized to regulate stem cell proliferation or lineage choice. However, the structure of adult neural stem cells and the contact they form with niche cells are poorly described. Here, we characterized the morphology of radial glia-like (RGL) cells, their molecular identity, proliferative activity, and fate determination in the adult mouse hippocampus. We found the coexistence of two morphotypes of cells with prototypical morphological characteristics of RGL stem cells: Type α cells, which represented 76% of all RGL cells, displayed a long primary process modestly branching into the molecular layer and type β cells, which represented 24% of all RGL cells, with a shorter radial process highly branching into the outer granule cell layer-inner molecular layer border. Stem cell markers were expressed in type α cells and coexpressed with astrocytic markers in type β cells. Consistently, in vivo lineage tracing indicated that type α cells can give rise to neurons, astrocytes, and type β cells, whereas type β cells do not proliferate. Our results reveal that the adult subgranular zone of the dentate gyrus harbors two functionally different RGL cells, which can be distinguished by simple morphological criteria, supporting a morphofunctional role of their thin cellular processes. Type β cells may represent an intermediate state in the transformation of type α, RGL stem cells, into astrocytes.
- Published
- 2016
191. Diversity of Neural Precursors in the Adult Mammalian Brain
- Author
-
Michael A. Bonaguidi, Guo Li Ming, Hongjun Song, Ryan P. Stadel, Jiaqi Sun, and Daniel A. Berg
- Subjects
0301 basic medicine ,Cell type ,media_common.quotation_subject ,Cellular differentiation ,Neurogenesis ,Cell lineage ,Biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,Neural Stem Cells ,Animals ,Humans ,Cell Lineage ,media_common ,Mammals ,Extramural ,Brain ,Cell Differentiation ,Mammalian brain ,Rats ,030104 developmental biology ,PERSPECTIVES ,Dentate Gyrus ,Neuroscience ,Diversity (politics) - Abstract
Aided by advances in technology, recent studies of neural precursor identity and regulation have revealed various cell types as contributors to ongoing cell genesis in the adult mammalian brain. Here, we use stem-cell biology as a framework to highlight the diversity of adult neural precursor populations and emphasize their hierarchy, organization, and plasticity under physiological and pathological conditions.
- Published
- 2016
192. Therapeutic targeting of oxygen-sensing prolyl hydroxylases abrogates ATF4-dependent neuronal death and improves outcomes after brain hemorrhage in several rodent models
- Author
-
Frederick Colbourne, Guo Li Ming, David W. Killilea, Giovanni Coppola, John W. Cave, Theodore R. Holman, Hongjun Song, Roseleen F. John, Irina G. Gazaryan, Guohua Xi, Sama F. Sleiman, Frederick R. Maxfield, Guo-Hua Fong, Dana Cruz, Carsten Culmsee, Ishraq Alim, Marina Demetriades, Soah J. Khim, Richard F. Keep, Timothy J Schallert, Cyrille C. Thinnes, Lewis B. Morgenstern, Ryan Tappero, Christopher J. Schofield, Tzu Lan Yeh, Sandra Neitemeier, Megan W. Bourassa, Saravanan S. Karuppagounder, Jian Zhong, Yijing Su, Sunghee Cho, and Rajiv R. Ratan
- Subjects
0301 basic medicine ,Iron ,Procollagen-Proline Dioxygenase ,Oxidative phosphorylation ,Pharmacology ,Iron Chelating Agents ,medicine.disease_cause ,Article ,Toxicology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Genes, Reporter ,In vivo ,Animals ,Protein Isoforms ,Medicine ,Molecular Targeted Therapy ,cardiovascular diseases ,Cells, Cultured ,Neurons ,Intracerebral hemorrhage ,chemistry.chemical_classification ,Cell Death ,business.industry ,ATF4 ,Brain ,Recovery of Function ,General Medicine ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Activating Transcription Factor 4 ,In vitro ,Rats ,Oxygen ,Disease Models, Animal ,Neuroprotective Agents ,030104 developmental biology ,Enzyme ,Gene Expression Regulation ,chemistry ,Hemin ,Liberation ,business ,Intracranial Hemorrhages ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
Disability or death due to intracerebral hemorrhage (ICH) is attributed to blood lysis, liberation of iron, and consequent oxidative stress. Iron chelators bind to free iron and prevent neuronal death induced by oxidative stress and disability due to ICH, but the mechanisms for this effect remain unclear. We show that the hypoxia-inducible factor prolyl hydroxylase domain (HIF-PHD) family of iron-dependent, oxygen-sensing enzymes are effectors of iron chelation. Molecular reduction of the three HIF-PHD enzyme isoforms in the mouse striatum improved functional recovery after ICH. A low-molecular-weight hydroxyquinoline inhibitor of the HIF-PHD enzymes, adaptaquin, reduced neuronal death and behavioral deficits after ICH in several rodent models without affecting total iron or zinc distribution in the brain. Unexpectedly, protection from oxidative death in vitro or from ICH in vivo by adaptaquin was associated with suppression of activity of the prodeath factor ATF4 rather than activation of an HIF-dependent prosurvival pathway. Together, these findings demonstrate that brain-specific inactivation of the HIF-PHD metalloenzymes with the blood-brain barrier–permeable inhibitor adaptaquin can improve functional outcomes after ICH in several rodent models.
- Published
- 2016
193. A unifying hypothesis on mammalian neural stem cell properties in the adult hippocampus
- Author
-
Juan Song, Guo Li Ming, Michael A. Bonaguidi, and Hongjun Song
- Subjects
Mammals ,Neurons ,Future studies ,Neurogenesis ,General Neuroscience ,Cellular differentiation ,Hippocampus ,Cell Differentiation ,Biology ,Article ,Neural stem cell ,Neuroepithelial cell ,Adult Stem Cells ,Cellular origin ,Animals ,Humans ,Neuroscience ,Adult stem cell - Abstract
Continuously generated new neurons promote circuitry plasticity within specialized regions and contribute to specific functions of the adult mammalian brain. A number of recent studies have investigated the cellular origin of adult neurogenesis in the hippocampus, yielding divergent models of neural stem cell behavior. An essential question remains whether these models are overlapping or fundamentally discrete. We review evidence that primary neural precursors in the adult hippocampus exhibit significant heterogeneity in their properties of self-renewal, multi-lineage differentiation and regulation, representing a range from unipotential committed precursors to bona fide self-renewing multipotent neural stem cells. We further present a testable unifying hypothesis of adult neural stem cell behavior in vivo to outline a common framework for future studies of molecular and cellular mechanisms regulating adult neural stem cells and how these cells may contribute to hippocampal function and repair.
- Published
- 2012
- Full Text
- View/download PDF
194. Modification of hippocampal circuitry by adult neurogenesis
- Author
-
Kimberly M. Christian, Hongjun Song, Guo Li Ming, and Juan Song
- Subjects
Neurons ,education.field_of_study ,Neurogenesis ,Dentate gyrus ,Population ,Biology ,Hippocampal formation ,Granule cell ,Hippocampus ,Article ,Neural stem cell ,Adult Stem Cells ,Cellular and Molecular Neuroscience ,medicine.anatomical_structure ,Neuropoiesis ,Neural Stem Cells ,Developmental Neuroscience ,Neuroplasticity ,medicine ,Animals ,Nerve Net ,education ,Neuroscience - Abstract
The adult hippocampus is one of the primary neural structures involved in memory formation. In addition to synapse-specific modifications thought to encode information at the sub-cellular level, changes in the intrahippocampal neuro-populational activity and dynamics at the circuit-level may contribute substantively to the functional capacity of this region. Within the hippocampus, the dentate gyrus has the potential to make a preferential contribution to neural circuit modification owing to the continuous addition of new granule cell population. The integration of newborn neurons into pre-existing circuitry is hypothesized to deliver a unique processing capacity, as opposed to merely replacing dying granule cells. Recent studies have begun to assess the impact of hippocampal neurogenesis by examining the extent to which adult-born neurons participate in hippocampal networks, including when newborn neurons become engaged in ongoing network activity and how they modulate circuit dynamics via their unique intrinsic physiological properties. Understanding the contributions of adult neurogenesis to hippocampal function will provide new insight into the fundamental aspects of brain plasticity, which can be used to guide therapeutic interventions to replace neural populations damaged by disease or injury.
- Published
- 2012
- Full Text
- View/download PDF
195. Interplay between DISC1 and GABA Signaling Regulates Neurogenesis in Mice and Risk for Schizophrenia
- Author
-
Ju Young Kim, David St Clair, Cindy Y. Liu, Xin Duan, Emer L. Feighery, Hongjun Song, Juan Song, Bai Lu, Guo Li Ming, Dan Rujescu, Fengyu Zhang, Joseph H. Callicott, Kimberly M. Christian, Daniel R. Weinberger, and Zhexing Wen
- Subjects
Sodium-Potassium-Chloride Symporters ,Neurogenesis ,Nerve Tissue Proteins ,Hippocampal formation ,General Biochemistry, Genetics and Molecular Biology ,gamma-Aminobutyric acid ,Article ,DISC1 ,Mice ,medicine ,Animals ,Solute Carrier Family 12, Member 2 ,PI3K/AKT/mTOR pathway ,gamma-Aminobutyric Acid ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Depolarization ,Anatomy ,Dendrites ,Hyperpolarization (biology) ,Mice, Inbred C57BL ,biology.protein ,Schizophrenia ,Female ,Disease Susceptibility ,Signal transduction ,Single-Cell Analysis ,Neuroscience ,medicine.drug ,Signal Transduction - Abstract
SummaryHow extrinsic stimuli and intrinsic factors interact to regulate continuous neurogenesis in the postnatal mammalian brain is unknown. Here we show that regulation of dendritic development of newborn neurons by Disrupted-in-Schizophrenia 1 (DISC1) during adult hippocampal neurogenesis requires neurotransmitter GABA-induced, NKCC1-dependent depolarization through a convergence onto the AKT-mTOR pathway. In contrast, DISC1 fails to modulate early-postnatal hippocampal neurogenesis when conversion of GABA-induced depolarization to hyperpolarization is accelerated. Extending the period of GABA-induced depolarization or maternal deprivation stress restores DISC1-dependent dendritic regulation through mTOR pathway during early-postnatal hippocampal neurogenesis. Furthermore, DISC1 and NKCC1 interact epistatically to affect risk for schizophrenia in two independent case control studies. Our study uncovers an interplay between intrinsic DISC1 and extrinsic GABA signaling, two schizophrenia susceptibility pathways, in controlling neurogenesis and suggests critical roles of developmental tempo and experience in manifesting the impact of susceptibility genes on neuronal development and risk for mental disorders.
- Published
- 2012
- Full Text
- View/download PDF
196. Time-dependent involvement of adult-born dentate granule cells in behavior
- Author
-
Woon Ryoung Kim, Guo Li Ming, Kimberly M. Christian, and Hongjun Song
- Subjects
Neurons ,Time Factors ,Behavior, Animal ,Recall ,Neurogenesis ,Dentate gyrus ,Granule (cell biology) ,Cognition ,Hippocampal function ,Hippocampal formation ,Article ,Behavioral Neuroscience ,Dentate Gyrus ,Animals ,Psychology ,Neuronal population ,Neuroscience - Abstract
Adult-born neurons are continuously generated and incorporated into the circuitry of the hippocampus throughout life in mammals. Cumulative evidence supports a physiological role for adult-born neurons, yet it not clear whether this subset of dentate granule cells makes a unique contribution to hippocampal function. Perturbation or ablation of adult hippocampal neurogenesis leads to deficits in the acquisition of learned associations or memory recall, whereas an increase in adult hippocampal neurogenesis enhances some forms of learning and memory. The observed effects thus far appear to be task-dependent, species-specific, and sensitive to the timing of manipulations. Here, we review the recent evidence correlating adult-born dentate granule cells (DGCs) with hippocampal-dependent behavior and focus on the dynamic properties of this neuronal population that may underlie its function. We further discuss a framework for future investigations of how newly integrated neurons may contribute to hippocampal processing using advanced genetic techniques with enhanced temporal resolution.
- Published
- 2012
- Full Text
- View/download PDF
197. 717. Using Human iPSCs for Psychiatric Disorder Disease Modeling and Mechanism-Based Drug Discovery
- Author
-
Kimberly M. Christian, Guo Li Ming, Wei Zheng, Namshik Kim, Ziyuan Guo, Zhexing Wen, Hongjun Song, Xinyuan Wang, Christopher A. Ross, Menghang Xia, Gong Chen, and Russell L. Margolis
- Subjects
0301 basic medicine ,Drug discovery ,business.industry ,Mechanism based ,Disease ,Bioinformatics ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Medicine ,Induced pluripotent stem cell ,business ,030217 neurology & neurosurgery ,Biological Psychiatry - Published
- 2017
- Full Text
- View/download PDF
198. Interaction between FEZ1 and DISC1 in Regulation of Neuronal Development and Risk for Schizophrenia
- Author
-
Ju Young Kim, Xin Duan, Junjie U. Guo, Anil K. Malhotra, Bai Lu, Guo Li Ming, Sungkyung Choi, Kimberly M. Christian, Katherine E. Burdick, Dimitrios Avramopoulos, Kurt A. Sailor, Eunchai Kang, Dhong Eun Jung, Dennis A. Pradhan, Hongjun Song, and Sundar Ganesan
- Subjects
Scaffold protein ,biology ,NDEL1 ,Neuroscience(all) ,General Neuroscience ,Neurogenesis ,Morphogenesis ,Hippocampus ,medicine.disease ,DISC1 ,Schizophrenia ,biology.protein ,medicine ,Neuroscience ,FEZ1 - Abstract
SummaryDisrupted-in Schizophrenia 1 (DISC1), a susceptibility gene for major mental disorders, encodes a scaffold protein that has a multifaceted impact on neuronal development. How DISC1 regulates different aspects of neuronal development is not well understood. Here, we show that Fasciculation and Elongation Protein Zeta-1 (FEZ1) interacts with DISC1 to synergistically regulate dendritic growth of newborn neurons in the adult mouse hippocampus, and that this pathway complements a parallel DISC1-NDEL1 interaction that regulates cell positioning and morphogenesis of newborn neurons. Furthermore, genetic association analysis of two independent cohorts of schizophrenia patients and healthy controls reveals an epistatic interaction between FEZ1 and DISC1, but not between FEZ1 and NDEL1, for risk of schizophrenia. Our findings support a model in which DISC1 regulates distinct aspects of neuronal development through its interaction with different intracellular partners and such epistasis may contribute to increased risk for schizophrenia.
- Published
- 2011
- Full Text
- View/download PDF
199. Emerging roles of TET proteins and 5-hydroxymethylcytosines in active DNA demethylation and beyond
- Author
-
Yijing Su, Junjie U. Guo, Guo Li Ming, Hongjun Song, and Chun Zhong
- Subjects
DNA Repair ,Protein family ,Cell Cycle Proteins ,Review ,Dioxygenases ,Mixed Function Oxygenases ,Cytosine ,chemistry.chemical_compound ,Proto-Oncogene Proteins ,Animals ,Humans ,Epigenetics ,Molecular Biology ,Genetics ,5-Hydroxymethylcytosine ,biology ,Nuclear Proteins ,Cell Biology ,DNA Methylation ,Cell biology ,DNA-Binding Proteins ,DNA demethylation ,chemistry ,DNA methylation ,5-Methylcytosine ,biology.protein ,Demethylase ,GADD45B ,DNA ,Developmental Biology - Abstract
Cytosine methylation is the major epigenetic modification of metazoan DNA. Although there is strong evidence that active DNA demethylation occurs in animal cells, the molecular details of this process are unknown. The recent discovery of the TET protein family (TET1-3) 5-methylcytosine hydroxylases has provided a new entry point to reveal the identity of the long-sought DNA demethylase. Here, we review the recent progress in understanding the function of TET proteins and 5-hydroxymethylcytosine (5hmC) through various biochemical and genomic approaches, the current evidence for a role of 5hmC as an early intermediate in active DNA demethylation and the potential functions of TET proteins and 5hmC beyond active DNA demethylation. We also discuss how future studies can extend our knowledge of this novel epigenetic modification.
- Published
- 2011
- Full Text
- View/download PDF
200. Neuronal activity modifies DNA methylation landscape in the adult brain
- Author
-
Hongjun Song, Jacob A. Balazer, Michael A. Bonaguidi, Bin Xie, Mi Hyeon Jang, Dengke K. Ma, Eric C. Ford, Yuan Gao, Guo Li Ming, Junjie U. Guo, Kun Zhang, Hugh L. Eaves, Huan Mo, and Madeleine Ball
- Subjects
Epigenomics ,Molecular Sequence Data ,Statistics as Topic ,Biology ,Motor Activity ,Hippocampus ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Epigenetics of physical exercise ,In vivo ,Physical Conditioning, Animal ,Premovement neuronal activity ,Animals ,030304 developmental biology ,Neurons ,0303 health sciences ,General Neuroscience ,Chromosome Mapping ,Genomics ,DNA Methylation ,Neuronal activation ,nervous system ,chemistry ,Gene Expression Regulation ,DNA methylation ,CpG Islands ,Neuroscience ,030217 neurology & neurosurgery ,DNA - Abstract
DNA methylation has been traditionally viewed as a highly stable epigenetic mark in post-mitotic cells, however, postnatal brains appear to exhibit stimulus-induced methylation changes, at least in a few identified CpG dinucleotides. How extensively the neuronal DNA methylome is regulated by neuronal activity is unknown. Using a next-generation sequencing-based method for genome-wide analysis at a single-nucleotide resolution, we quantitatively compared the CpG methylation landscape of adult mouse dentate granule neurons in vivo before and after synchronous neuronal activation. About 1.4% of 219,991 CpGs measured show rapid active demethylation or de novo methylation. Some modifications remain stable for at least 24 hours. These activity-modified CpGs exhibit a broad genomic distribution with significant enrichment in low-CpG density regions, and are associated with brain-specific genes related to neuronal plasticity. Our study implicates modification of the neuronal DNA methylome as a previously under-appreciated mechanism for activity-dependent epigenetic regulation in the adult nervous system.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.