Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cells ; 13(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667322

ABSTRACT

Although lineage reprogramming from one cell type to another is becoming a breakthrough technology for cell-based therapy, several limitations remain to be overcome, including the low conversion efficiency and subtype specificity. To address these, many studies have been conducted using genetics, chemistry, physics, and cell biology to control transcriptional networks, signaling cascades, and epigenetic modifications during reprogramming. Here, we summarize recent advances in cellular reprogramming and discuss future directions.


Subject(s)
Cell Lineage , Cellular Reprogramming , Cellular Reprogramming/genetics , Humans , Animals , Cell Lineage/genetics , Neurons/metabolism , Neurons/cytology , Epigenesis, Genetic , Cell Differentiation/genetics , Signal Transduction
2.
Proc Natl Acad Sci U S A ; 120(42): e2307972120, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37812721

ABSTRACT

Although generating new neurons in the ischemic injured brain would be an ideal approach to replenish the lost neurons for repairing the damage, the adult mammalian brain retains only limited neurogenic capability. Here, we show that direct conversion of microglia/macrophages into neurons in the brain has great potential as a therapeutic strategy for ischemic brain injury. After transient middle cerebral artery occlusion in adult mice, microglia/macrophages converge at the lesion core of the striatum, where neuronal loss is prominent. Targeted expression of a neurogenic transcription factor, NeuroD1, in microglia/macrophages in the injured striatum enables their conversion into induced neuronal cells that functionally integrate into the existing neuronal circuits. Furthermore, NeuroD1-mediated induced neuronal cell generation significantly improves neurological function in the mouse stroke model, and ablation of these cells abolishes the gained functional recovery. Our findings thus demonstrate that neuronal conversion contributes directly to functional recovery after stroke.


Subject(s)
Brain Ischemia , Stroke , Mice , Animals , Microglia/metabolism , Stroke/metabolism , Macrophages/metabolism , Brain/metabolism , Neurons/metabolism , Brain Ischemia/metabolism , Infarction, Middle Cerebral Artery/metabolism , Mammals
3.
Genes Cells ; 28(7): 526-534, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37114566

ABSTRACT

Neuronal regeneration to replenish lost neurons after injury is critical for brain repair. Microglia, brain-resident macrophages that have the propensity to accumulate at the site of injury, can be a potential source for replenishing lost neurons through fate conversion into neurons, induced by forced expression of neuronal lineage-specific transcription factors. However, it has not been strictly demonstrated that microglia, rather than central nervous system-associated macrophages, such as meningeal macrophages, convert into neurons. Here, we show that NeuroD1-transduced microglia can be successfully converted into neurons in vitro using lineage-mapping strategies. We also found that a chemical cocktail treatment further promoted NeuroD1-induced microglia-to-neuron conversion. NeuroD1 with loss-of-function mutation, on the other hand, failed to induce the neuronal conversion. Our results indicate that microglia are indeed reprogrammed into neurons by NeuroD1 with neurogenic transcriptional activity.


Subject(s)
Microglia , Neurons , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/metabolism , Microglia/metabolism , Neurogenesis , Neurons/metabolism , Transcription Factors/metabolism , Animals , Mice
4.
Sci Rep ; 12(1): 17980, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36289433

ABSTRACT

Several transcription factors, including NeuroD1, have been shown to act as neuronal reprogramming factors (RFs) that induce neuronal conversion from somatic cells. However, it remains unexplored whether expression levels of RFs in the original cells affect reprogramming efficiency. Here, we show that the neuronal reprogramming efficiency from two distinct glial cell types, microglia and astrocytes, is substantially dependent on the expression level of NeuroD1: low expression failed to induce neuronal reprogramming, whereas elevated NeuroD1 expression dramatically improved reprogramming efficiency in both cell types. Moreover, even under conditions where NeuroD1 expression was too low to induce effective conversion by itself, combined expression of three RFs (Ascl1, Brn2, and NeuroD1) facilitated the breaking down of cellular barriers, inducing neuronal reprogramming. Thus, our results suggest that a sufficiently high expression level of RFs, or alternatively their combinatorial expression, is the key to achieving efficient neuronal reprogramming from different cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cellular Reprogramming , Cellular Reprogramming/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Neurons/metabolism , Astrocytes/metabolism , Transcription Factors/metabolism , Neuroglia/metabolism
6.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Article in English | MEDLINE | ID: mdl-34526402

ABSTRACT

Linkage between early-life exposure to anesthesia and subsequent learning disabilities is of great concern to children and their families. Here we show that early-life exposure to midazolam (MDZ), a widely used drug in pediatric anesthesia, persistently alters chromatin accessibility and the expression of quiescence-associated genes in neural stem cells (NSCs) in the mouse hippocampus. The alterations led to a sustained restriction of NSC proliferation toward adulthood, resulting in a reduction of neurogenesis that was associated with the impairment of hippocampal-dependent memory functions. Moreover, we found that voluntary exercise restored hippocampal neurogenesis, normalized the MDZ-perturbed transcriptome, and ameliorated cognitive ability in MDZ-exposed mice. Our findings thus explain how pediatric anesthesia provokes long-term adverse effects on brain function and provide a possible therapeutic strategy for countering them.


Subject(s)
Chromatin/drug effects , Midazolam/adverse effects , Neurogenesis/drug effects , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chromatin/metabolism , Cognition/drug effects , Cognition/physiology , Female , Hippocampus/drug effects , Hippocampus/metabolism , Male , Memory , Mice , Mice, Inbred C57BL , Midazolam/pharmacology , Models, Animal , Neural Stem Cells/metabolism , Neurogenesis/physiology
7.
Cells ; 10(5)2021 05 10.
Article in English | MEDLINE | ID: mdl-34068607

ABSTRACT

Tissue-specific stem cells give rise to new functional cells to maintain tissue homeostasis and restore damaged tissue after injury. To ensure proper brain functions in the adult brain, neural stem cells (NSCs) continuously generate newborn neurons that integrate into pre-existing neuronal networks. Proliferation, as well as neurogenesis of NSCs, are exquisitely controlled by extrinsic and intrinsic factors, and their underlying mechanisms have been extensively studied with the goal of enhancing the neurogenic capacity of NSCs for regenerative medicine. However, neurogenesis of endogenous NSCs alone is insufficient to completely repair brains damaged by neurodegenerative diseases and/or injury because neurogenic areas are limited and few neurons are produced in the adult brain. An innovative approach towards replacing damaged neurons is to induce conversion of non-neuronal cells residing in injured sites into neurons by a process referred to as direct reprogramming. This review describes extrinsic and intrinsic factors controlling NSCs and neurogenesis in the adult brain and discusses prospects for their applications. It also describes direct neuronal reprogramming technology holding promise for future clinical applications.


Subject(s)
Adult Stem Cells/physiology , Brain/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Animals , Astrocytes/cytology , Cell Differentiation/physiology , Cell Lineage , Cell Proliferation , Extracellular Matrix/metabolism , Hippocampus/metabolism , Humans , Mice , Microglia , Nerve Net , Neurodegenerative Diseases , Neurons/metabolism , Neurons/physiology , Regenerative Medicine , Signal Transduction , Stem Cell Niche/physiology , Transcriptome
9.
Neurosci Res ; 166: 1-11, 2021 May.
Article in English | MEDLINE | ID: mdl-32497571

ABSTRACT

Neural stem cells (NSCs) in the adult hippocampus generate new neurons via a process referred to as neurogenesis, supporting cognitive functions. Since altered neurogenesis has been reportedly associated with several diseases such as epilepsy, the molecular basis of NSC activity is an important focus in the study of neurogenesis. Furthermore, facilitation of neurogenesis in the injured brain would be an ideal approach to replenish lost neurons for damage recovery. However, natural neurogenesis by endogenous NSCs in the adult brain is insufficient for complete recovery after severe injury. Recent advances in understanding forced neurogenesis from brain-resident non-neuronal cells by direct reprogramming and clearing hurdles to achieve it have improved the ability to replace damaged neurons in the brain. In this review, we describe molecular mechanisms underlying natural and forced neurogenesis, and discuss future directions for treatments of diseases in the central nervous system.


Subject(s)
Neural Stem Cells , Neurogenesis , Brain , Hippocampus , Neurons
10.
Neuron ; 101(3): 472-485.e7, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30638745

ABSTRACT

Minimal sets of transcription factors can directly reprogram somatic cells into neurons. However, epigenetic remodeling during neuronal reprogramming has not been well reconciled with transcriptional regulation. Here we show that NeuroD1 achieves direct neuronal conversion from mouse microglia both in vitro and in vivo. Exogenous NeuroD1 initially occupies closed chromatin regions associated with bivalent trimethylation of histone H3 at lysine 4 (H3K4me3) and H3K27me3 marks in microglia to induce neuronal gene expression. These regions are resolved to a monovalent H3K4me3 mark at later stages of reprogramming to establish the neuronal identity. Furthermore, the transcriptional repressors Scrt1 and Meis2 are induced as NeuroD1 target genes, resulting in a decrease in the expression of microglial genes. In parallel, the microglial epigenetic signature in promoter and enhancer regions is erased. These findings reveal NeuroD1 pioneering activity accompanied by global epigenetic remodeling for two sequential events: onset of neuronal property acquisition and loss of the microglial identity during reprogramming.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cellular Reprogramming , Epigenesis, Genetic , Microglia/cytology , Neurons/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Corpus Striatum/cytology , Female , HEK293 Cells , Histone Code , Histones/chemistry , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Microglia/metabolism , Neurons/metabolism
11.
Neurosci Res ; 143: 31-43, 2019 Jun.
Article in English | MEDLINE | ID: mdl-29859850

ABSTRACT

Adult neurogenesis is a process of generating new neurons from neural stem/precursor cells (NS/PCs) in restricted adult brain regions throughout life. It is now generally known that adult neurogenesis in the hippocampal dentate gyrus (DG) and subventricular zone participates in various higher brain functions, such as learning and memory formation, olfactory discrimination and repair after brain injury. However, the mechanisms underlying adult neurogenesis remain to be fully understood. Here, we show that Nuclear protein 95 KDa (Np95, also known as UHRF1 or ICBP90), which is an essential protein for maintaining DNA methylation during cell division, is involved in multiple processes of adult neurogenesis. Specific ablation of Np95 in adult NS/PCs (aNS/PCs) led to a decrease in their proliferation and an impairment of neuronal differentiation and to suppression of neuronal maturation associated with the impairment of dendritic formation in the hippocampal DG. We also found that deficiency of Np95 in NS/PCs increased the expression of tumor suppressor genes p16 and p53, and confirmed that expression of these genes in NS/PCs recapitulates the phenotype of Np95-deficient NS/PCs. Taken together, our findings suggest that Np95 plays an essential role in proliferation and differentiation of aNS/PCs through the regulation of tumor suppressor gene expression in adult neurogenesis.


Subject(s)
Adult Stem Cells/physiology , Gene Expression Regulation , Genes, Tumor Suppressor , Neural Stem Cells/physiology , Nuclear Proteins/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , CCAAT-Enhancer-Binding Proteins , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Dentate Gyrus/metabolism , Hippocampus/metabolism , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Nuclear Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases
12.
Proc Natl Acad Sci U S A ; 115(16): 4270-4275, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610328

ABSTRACT

Epilepsy is a neurological disorder often associated with seizure that affects ∼0.7% of pregnant women. During pregnancy, most epileptic patients are prescribed antiepileptic drugs (AEDs) such as valproic acid (VPA) to control seizure activity. Here, we show that prenatal exposure to VPA in mice increases seizure susceptibility in adult offspring through mislocalization of newborn neurons in the hippocampus. We confirmed that neurons newly generated from neural stem/progenitor cells (NS/PCs) are integrated into the granular cell layer in the adult hippocampus; however, prenatal VPA treatment altered the expression in NS/PCs of genes associated with cell migration, including CXC motif chemokine receptor 4 (Cxcr4), consequently increasing the ectopic localization of newborn neurons in the hilus. We also found that voluntary exercise in a running wheel suppressed this ectopic neurogenesis and countered the enhanced seizure susceptibility caused by prenatal VPA exposure, probably by normalizing the VPA-disrupted expression of multiple genes including Cxcr4 in adult NS/PCs. Replenishing Cxcr4 expression alone in NS/PCs was sufficient to overcome the aberrant migration of newborn neurons and increased seizure susceptibility in VPA-exposed mice. Thus, prenatal exposure to an AED, VPA, has a long-term effect on the behavior of NS/PCs in offspring, but this effect can be counteracted by a simple physical activity. Our findings offer a step to developing strategies for managing detrimental effects in offspring exposed to VPA in utero.


Subject(s)
Anticonvulsants/toxicity , Neurogenesis/drug effects , Prenatal Exposure Delayed Effects , Seizures/etiology , Valproic Acid/toxicity , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/pharmacology , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Dentate Gyrus/drug effects , Dentate Gyrus/embryology , Dentate Gyrus/pathology , Disease Susceptibility , Female , Gene Expression Regulation, Developmental/drug effects , Gestational Age , Hippocampus/embryology , Hippocampus/pathology , Hippocampus/physiopathology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neural Stem Cells/drug effects , Neurons/pathology , Physical Exertion , Pregnancy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Rats , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/genetics , Receptors, CXCR4/therapeutic use , Seizures/chemically induced , Seizures/embryology , Transcriptome , Valproic Acid/administration & dosage , Valproic Acid/pharmacology
13.
Stem Cells ; 36(5): 737-750, 2018 05.
Article in English | MEDLINE | ID: mdl-29517828

ABSTRACT

Together with residual host neurons, transplanted neural stem cell (NSC)-derived neurons play a critical role in reconstructing disrupted neural circuits after spinal cord injury (SCI). Since a large number of tracts are disrupted and the majority of host neurons die around the lesion site as the damage spreads, minimizing this spreading and preserving the lesion site are important for attaining further improvements in reconstruction. High mobility group box-1 (HMGB1) is a damage-associated molecular pattern protein that triggers sterile inflammation after tissue injury. In the ischemic and injured brain, neutralization of HMGB1 with a specific antibody reportedly stabilizes the blood-brain barrier, suppresses inflammatory cytokine expression, and improves functional recovery. Using a SCI model mouse, we here developed a combinatorial treatment for SCI: administering anti-HMGB1 antibody prior to transplantation of NSCs derived from human induced pluripotent stem cells (hiPSC-NSCs) yielded a dramatic improvement in locomotion recovery after SCI. Even anti-HMGB1 antibody treatment alone alleviated blood-spinal cord barrier disruption and edema formation, and increased the number of neurites from spared axons and the survival of host neurons, resulting in functional recovery. However, this recovery was greatly enhanced by the subsequent hiPSC-NSC transplantation, reaching an extent that has never before been reported. We also found that this improved recovery was directly associated with connections established between surviving host neurons and transplant-derived neurons. Taken together, our results highlight combinatorial treatment with anti-HMGB1 antibody and hiPSC-NSC transplantation as a promising novel therapy for SCI. Stem Cells 2018;36:737-750.


Subject(s)
Cell Differentiation/physiology , Neural Stem Cells/cytology , Recovery of Function/physiology , Spinal Cord Injuries/therapy , Animals , Cells, Cultured , Disease Models, Animal , HMGB1 Protein/immunology , Humans , Mice, Inbred NOD , Mice, SCID , Stem Cell Transplantation/methods
14.
Dev Dyn ; 247(1): 229-238, 2018 01.
Article in English | MEDLINE | ID: mdl-28771884

ABSTRACT

BACKGROUND: Although quiescent neural stem cells (NSCs) in the adult hippocampus proliferate in response to neurogenic stimuli and subsequently give rise to new neurons continuously throughout life, misregulation of NSCs in pathological conditions, including aging, leads to the impairment of learning and memory. High mobility group B family 1 (HMGB1) and HMGB2, HMG family proteins that function as transcriptional activators through the modulation of chromatin structure, have been assumed to play some role in the regulation of adult NSCs; however, their precise functions and even expression patterns in the adult hippocampus remain elusive. RESULTS: Here we show that expression of HMGB2 but not HMGB1 is restricted to the subset of NSCs and their progenitors. Furthermore, running, a well-known positive neurogenic stimulus, increased the proliferation of HMGB2-expressing cells, whereas aging was accompanied by a marked decrease in these cells. Intriguingly, HMGB2-expressing quiescent NSCs, which were shifted toward the proliferative state, were decreased as aging progressed. CONCLUSIONS: HMGB2 expression is strongly associated with transition from the quiescent to the proliferative state of NSCs, supporting the possibility that HMGB2 is involved in the regulation of adult neurogenesis and can be used as a novel marker to identify NSCs primed for activation in the adult hippocampus. Developmental Dynamics 247:229-238, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Adult Stem Cells/metabolism , HMGB2 Protein/metabolism , Hippocampus/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Adult Stem Cells/cytology , Animals , HMGB2 Protein/genetics , Hippocampus/cytology , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Mice , Neural Stem Cells/cytology , Neurons/cytology , Neurons/metabolism
15.
Stem Cell Reports ; 8(6): 1743-1756, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28591654

ABSTRACT

Human neural precursor cells (hNPCs) derived from pluripotent stem cells display a high propensity for neuronal differentiation, but they require long-term culturing to differentiate efficiently into astrocytes. The mechanisms underlying this biased fate specification of hNPCs remain elusive. Here, we show that hypoxia confers astrocytic differentiation potential on hNPCs through epigenetic gene regulation, and that this was achieved by cooperation between hypoxia-inducible factor 1α and Notch signaling, accompanied by a reduction of DNA methylation level in the promoter region of a typical astrocyte-specific gene, Glial fibrillary acidic protein. Furthermore, we found that this hypoxic culture condition could be applied to rapid generation of astrocytes from Rett syndrome patient-derived hNPCs, and that these astrocytes impaired neuronal development. Thus, our findings shed further light on the molecular mechanisms regulating hNPC differentiation and provide attractive tools for the development of therapeutic strategies for treating astrocyte-mediated neurological disorders.


Subject(s)
Astrocytes/cytology , Cell Differentiation , Cell Hypoxia , Epigenomics , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Astrocytes/metabolism , Binding Sites , Cell Line , CpG Islands , DNA Methylation , Glial Fibrillary Acidic Protein/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neural Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Receptors, Notch/metabolism , Rett Syndrome/metabolism , Rett Syndrome/pathology , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/metabolism , Signal Transduction
16.
Stem Cell Reports ; 8(6): 1506-1515, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28506534

ABSTRACT

Currently, all methods for converting non-neuronal cells into neurons involve injury to the brain; however, whether neuronal transdifferentiation can occur long after the period of insult remains largely unknown. Here, we use the transcription factor NEUROD1, previously shown to convert reactive glial cells to neurons in the cortex, to determine whether astrocyte-to-neuron transdifferentiation can occur under physiological conditions. We utilized adeno-associated virus 9 (AAV9), which crosses the blood-brain barrier without injury, to deliver NEUROD1 to astrocytes through an intravascular route. Interestingly, we found that a small, but significant number of non-reactive astrocytes converted to neurons in the striatum, but not the cortex. Moreover, astrocytes cultured to minimize their proliferative potential also exhibited limited neuronal transdifferentiation with NEUROD1 expression. Our results show that a single transcription factor can induce astrocyte-to-neuron conversion under physiological conditions, potentially facilitating future clinical approaches long after the acute injury phase.


Subject(s)
Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Astrocytes/cytology , Basic Helix-Loop-Helix Transcription Factors/genetics , Blood-Brain Barrier/metabolism , Cell Differentiation , Cell Transdifferentiation , Cells, Cultured , Cellular Reprogramming , Corpus Striatum/metabolism , Dependovirus/genetics , Genetic Vectors/genetics , Genetic Vectors/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/cytology , Rats
17.
J Neurosci ; 36(22): 6050-68, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27251626

ABSTRACT

UNLABELLED: Development of the hippocampal dentate gyrus (DG) in the mammalian brain is achieved through multiple processes during late embryonic and postnatal stages, with each developmental step being strictly governed by extracellular cues and intracellular mechanisms. Here, we show that the maintenance DNA methyltransferase 1 (Dnmt1) is critical for development of the DG in the mouse. Deletion of Dnmt1 in neural stem cells (NSCs) at the beginning of DG development led to a smaller size of the granule cell layer in the DG. NSCs lacking Dnmt1 failed to establish proper radial processes or to migrate into the subgranular zone, resulting in aberrant neuronal production in the molecular layer of the DG and a reduction of integrated neurons in the granule cell layer. Interestingly, prenatal deletion of Dnmt1 in NSCs affected not only the developmental progression of the DG but also the properties of NSCs maintained into adulthood: Dnmt1-deficient NSCs displayed impaired neurogenic ability and proliferation. We also found that Dnmt1 deficiency in NSCs decreased the expression of Reelin signaling components in the developing DG and increased that of the cell cycle inhibitors p21 and p57 in the adult DG. Together, these findings led us to propose that Dnmt1 functions as a key regulator to ensure the proper development of the DG, as well as the proper status of NSCs maintained into adulthood, by modulating extracellular signaling and intracellular mechanisms. SIGNIFICANCE STATEMENT: Here, we provide evidence that Dnmt1 is required for the proper development of the hippocampal dentate gyrus (DG). Deletion of Dnmt1 in neural stem cells (NSCs) at an early stage of DG development impaired the ability of NSCs to establish secondary radial glial scaffolds and to migrate into the subgranular zone of the DG, leading to aberrant neuronal production in the molecular layer, increased cell death, and decreased granule neuron production. Prenatal deletion of Dnmt1 in NSCs also induced defects in the proliferation and neurogenic ability of adult NSCs. Furthermore, we found that Dnmt1 regulates the expression of key extracellular signaling components during developmental stages while modulating intracellular mechanisms for proliferation and neuronal production of NSCs in the adult.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Dentate Gyrus , Gene Expression Regulation, Developmental , Neurons/physiology , Animals , Animals, Newborn , Cell Differentiation/genetics , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1 , Dentate Gyrus/cytology , Dentate Gyrus/embryology , Dentate Gyrus/growth & development , Doublecortin Domain Proteins , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred ICR , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Neural Stem Cells/physiology , Neurogenesis/genetics , Neuropeptides/genetics , Neuropeptides/metabolism , Reelin Protein
18.
Nat Commun ; 6: 6514, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25751136

ABSTRACT

Pathological conditions such as epilepsy cause misregulation of adult neural stem/progenitor populations in the adult hippocampus in mice, and the resulting abnormal neurogenesis leads to impairment in learning and memory. However, how animals cope with abnormal neurogenesis remains unknown. Here we show that microglia in the mouse hippocampus attenuate convulsive seizure-mediated aberrant neurogenesis through the activation of Toll-like receptor 9 (TLR9), an innate immune sensor known to recognize microbial DNA and trigger inflammatory responses. We found that microglia sense self-DNA from degenerating neurons following seizure, and secrete tumour necrosis factor-α, resulting in attenuation of aberrant neurogenesis. Furthermore, TLR9 deficiency exacerbated seizure-induced cognitive decline and recurrent seizure severity. Our findings thus suggest the existence of bidirectional communication between the innate immune and nervous systems for the maintenance of adult brain integrity.


Subject(s)
Cognitive Dysfunction/immunology , Hippocampus/immunology , Microglia/immunology , Neurogenesis/immunology , Seizures/immunology , Toll-Like Receptor 9/genetics , Animals , Cognition , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Gene Expression Regulation , Hippocampus/growth & development , Hippocampus/metabolism , Hippocampus/pathology , Immunity, Innate/genetics , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Neurogenesis/genetics , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Seizures/genetics , Seizures/metabolism , Seizures/pathology , Severity of Illness Index , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
19.
Neurosci Res ; 95: 1-11, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25659757

ABSTRACT

Adult neurogenesis persists throughout life in the dentate gyrus (DG) of the hippocampus, and its importance has been highlighted in hippocampus-dependent learning and memory. Adult neurogenesis consists of multiple processes: maintenance and neuronal differentiation of neural stem/precursor cells (NS/PCs), followed by survival and maturation of newborn neurons and their integration into existing neuronal circuitry. However, the mechanisms that govern these processes remain largely unclear. Here we show that DNA methyltransferase 1 (DNMT1), an enzyme responsible for the maintenance of DNA methylation, is highly expressed in proliferative cells in the adult DG and plays an important role in the survival of newly generated neurons. Deletion of Dnmt1 in adult NS/PCs (aNS/PCs) did not affect the proliferation and differentiation of aNS/PCs per se. However, it resulted in a decrease of newly generated mature neurons, probably due to gradual cell death after aNS/PCs differentiated into neurons in the hippocampus. Interestingly, loss of DNMT1 in post-mitotic neurons did not influence their survival. Taken together, these findings suggest that the presence of DNMT1 in aNS/PCs is crucial for the survival of newly generated neurons, but is dispensable once they accomplish neuronal differentiation in the adult hippocampus.


Subject(s)
Adult Stem Cells/enzymology , DNA (Cytosine-5-)-Methyltransferases/metabolism , Dentate Gyrus/enzymology , Neural Stem Cells/enzymology , Neurons/enzymology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Dentate Gyrus/cytology , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Neurons/cytology
20.
Neurogenesis (Austin) ; 2(1): e1081714, 2015.
Article in English | MEDLINE | ID: mdl-27604264

ABSTRACT

A population of proliferating neural stem/progenitor cells located in the subgranular zone of the adult hippocampal dentate gyrus (DG) gives rise to new neurons continuously throughout life, and this process is referred to as adult hippocampal neurogenesis. To date, it has generally been accepted that impairments of adult hippocampal neurogenesis resulting from pathological conditions such as stress, ischemia and epilepsy lead to deficits in hippocampus-dependent learning and memory tasks. Recently, we have discovered that microglia, the major immune cells in the brain, attenuate seizure-induced aberrant hippocampal neurogenesis to withstand cognitive decline and recurrent seizure. In that study, we further showed that Toll-like receptor 9, known as a pathogen-sensing receptor for innate immune system activation, recognizes self-DNA derived from degenerating neurons to induce TNF-α production in the microglia after seizure, resulting in inhibition of seizure-induced aberrant neurogenesis. Our findings provide new evidence that interaction between the innate immune and nervous systems ensures homeostatic neurogenesis in the adult hippocampus and should pave the way for the development of new therapeutic strategies for neurological diseases including epilepsy.

SELECTION OF CITATIONS
SEARCH DETAIL
...