Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 223(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38652118

ABSTRACT

Chromatin-remodeling protein BRG1/SMARCA4 is pivotal for establishing oligodendrocyte (OL) lineage identity. However, its functions for oligodendrocyte-precursor cell (OPC) differentiation within the postnatal brain and during remyelination remain elusive. Here, we demonstrate that Brg1 loss profoundly impairs OPC differentiation in the brain with a comparatively lesser effect in the spinal cord. Moreover, BRG1 is critical for OPC remyelination after injury. Integrative transcriptomic/genomic profiling reveals that BRG1 exhibits a dual role by promoting OPC differentiation networks while repressing OL-inhibitory cues and proneuronal programs. Furthermore, we find that BRG1 interacts with EED/PRC2 polycomb-repressive-complexes to enhance H3K27me3-mediated repression at gene loci associated with OL-differentiation inhibition and neurogenesis. Notably, BRG1 depletion decreases H3K27me3 deposition, leading to the upregulation of BMP/WNT signaling and proneurogenic genes, which suppresses OL programs. Thus, our findings reveal a hitherto unexplored spatiotemporal-specific role of BRG1 for OPC differentiation in the developing CNS and underscore a new insight into BRG1/PRC2-mediated epigenetic regulation that promotes and safeguards OL lineage commitment and differentiation.


Subject(s)
Cell Differentiation , DNA Helicases , Oligodendroglia , Polycomb Repressive Complex 2 , Animals , Mice , DNA Helicases/metabolism , DNA Helicases/genetics , Epigenesis, Genetic , Histones/metabolism , Histones/genetics , Mice, Inbred C57BL , Neurogenesis/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/genetics , Remyelination , Transcription Factors/metabolism , Transcription Factors/genetics
2.
Nat Commun ; 14(1): 2186, 2023 04 17.
Article in English | MEDLINE | ID: mdl-37069258

ABSTRACT

Despite extensive research on astrocytic Ca2+ in synaptic transmission, its contribution to the modulation of sensory transmission during different brain states remains largely unknown. Here, by using two-photon microscopy and whole-cell recordings, we show two distinct astrocytic Ca2+ signals in the murine barrel cortex: a small, long-lasting Ca2+ increase during sleep and a large, widespread but short-lasting Ca2+ spike when aroused. The large Ca2+ wave in aroused mice was inositol trisphosphate (IP3)-dependent, evoked by the locus coeruleus-norepinephrine system, and enhanced sensory input, contributing to reliable sensory transmission. However, the small Ca2+ transient was IP3-independent and contributed to decreased extracellular K+, hyperpolarization of the neurons, and suppression of sensory transmission. These events respond to different pharmacological inputs and contribute to distinct sleep and arousal functions by modulating the efficacy of sensory transmission. Together, our data demonstrate an important function for astrocytes in sleep and arousal states via astrocytic Ca2+ waves.


Subject(s)
Astrocytes , Wakefulness , Mice , Animals , Astrocytes/physiology , Calcium Signaling/physiology , Arousal/physiology , Sleep
3.
J Immunother Cancer ; 10(11)2022 11.
Article in English | MEDLINE | ID: mdl-36427906

ABSTRACT

BACKGROUND: Cancer immunotherapy has taken center stage in cancer treatment. However, the current immunotherapies only benefit a small proportion of patients with cancer, necessitating better understanding of the mechanisms of tumor immune evasion and improved cancer immunotherapy strategies. Regulatory T (Treg) cells play an important role in maintaining immune tolerance through inhibiting effector T-cell function. In the tumor microenvironment, Treg cells are used by tumor cells to counteract effector T cell-mediated tumor suppression. Targeting Treg cells may thus unleash the antitumor activity of effector T cells. While systemic depletion of Treg cells can cause excessive effector T-cell responses and subsequent autoimmune diseases, controlled targeting of Treg cells may benefit patients with cancer. METHODS: Treg cells from Treg cell-specific heterozygous Cdc42 knockout mice, C57BL/6 mice treated with a Cdc42 inhibitor CASIN, and control mice were examined for their homeostasis and stability by flow cytometry. The autoimmune responses in Treg cell-specific heterozygous Cdc42 knockout mice, CASIN-treated C57BL/6 mice, and control mice were assessed by H&E staining and ELISA. Antitumor T-cell immunity in Treg cell-specific heterozygous Cdc42 knockout mice, CASIN-treated C57BL/6 mice, humanized NSGS mice, and control mice was assessed by challenging the mice with MC38 mouse colon cancer cells, KPC mouse pancreatic cancer cells, or HCT116 human colon cancer cells. RESULTS: Treg cell-specific heterozygous deletion or pharmacological targeting of Cdc42 with CASIN does not affect Treg cell numbers but induces Treg cell instability, leading to antitumor T-cell immunity without detectable autoimmune reactions. Cdc42 targeting causes an additive effect on immune checkpoint inhibitor anti-programmed cell death protein-1 antibody-induced T-cell response against mouse and human tumors. Mechanistically, Cdc42 targeting induces Treg cell instability and unleashes antitumor T-cell immunity through carbonic anhydrase I-mediated pH changes. CONCLUSIONS: Rational targeting of Cdc42 in Treg cells holds therapeutic promises in cancer immunotherapy.


Subject(s)
Colonic Neoplasms , T-Lymphocytes, Regulatory , Humans , Mice , Animals , Mice, Inbred C57BL , Immunotherapy , Mice, Knockout , Tumor Microenvironment
4.
Neuron ; 110(20): 3318-3338.e9, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36265442

ABSTRACT

Brain tissue transcriptomes may be organized into gene coexpression networks, but their underlying biological drivers remain incompletely understood. Here, we undertook a large-scale transcriptomic study using 508 wild-type mouse striatal tissue samples dissected exclusively in the afternoons to define 38 highly reproducible gene coexpression modules. We found that 13 and 11 modules are enriched in cell-type and molecular complex markers, respectively. Importantly, 18 modules are highly enriched in daily rhythmically expressed genes that peak or trough with distinct temporal kinetics, revealing the underlying biology of striatal diurnal gene networks. Moreover, the diurnal coexpression networks are a dominant feature of daytime transcriptomes in the mouse cortex. We next employed the striatal coexpression modules to decipher the striatal transcriptomic signatures from Huntington's disease models and heterozygous null mice for 52 genes, uncovering novel functions for Prkcq and Kdm4b in oligodendrocyte differentiation and bipolar disorder-associated Trank1 in regulating anxiety-like behaviors and nocturnal locomotion.


Subject(s)
Huntington Disease , Transcriptome , Animals , Mice , Protein Kinase C-theta/genetics , Gene Regulatory Networks , Huntington Disease/genetics , Brain
5.
Front Endocrinol (Lausanne) ; 12: 698115, 2021.
Article in English | MEDLINE | ID: mdl-34646232

ABSTRACT

Previous studies have shown that agonists of GPR17 stimulate, while antagonists inhibit feeding. However, whole body knockout of GPR17 in mice of the C57Bl/6 strain did not affect energy balance, whereas selective knockout in oligodendrocytes or pro-opiomelanocortin neurons provided protection from high fat diet-induced obesity and impaired glucose homeostasis. We reasoned that whole body knockout of GPR17 in mice of the 129 strain might elicit more marked effects because the 129 strain is more susceptible than the C57Bl/6 strain to increased sympathetic activity and less susceptible to high fat diet-induced obesity. Consistent with this hypothesis, compared to wild-type mice, and when fed on either a chow or a high fat diet, GPR17 -/- mice of the 129 strain displayed increased expression of uncoupling protein-1 in white adipose tissue, lower body weight and fat content, reduced plasma leptin, non-esterified fatty acids and triglycerides, and resistance to high fat diet-induced glucose intolerance. Not only energy expenditure, but also energy intake was raised. Administration of leptin did not suppress the increased food intake in GPR17 -/- mice of the 129 strain, whereas it did suppress food intake in GPR17 +/+ mice. The only difference between GPR17 +/- and GPR17 +/+ mice of the C57Bl/6 strain was that the body weight of the GPR17 -/- mice was lower than that of the GPR17 +/+ mice when the mice were fed on a standard chow diet. We propose that the absence of GPR17 raises sympathetic activity in mice of the 129 strain in response to a low plasma fuel supply, and that the consequent loss of body fat is partly mitigated by increased energy intake.


Subject(s)
Energy Intake , Leptin/blood , Leptin/pharmacology , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Thinness/genetics , Adipose Tissue/metabolism , Animals , Body Composition/genetics , Energy Intake/drug effects , Energy Intake/physiology , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Species Specificity , Thinness/blood
6.
Neuro Oncol ; 23(7): 1183-1196, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33539525

ABSTRACT

BACKGROUND: Diffuse intrinsic pontine glioma (DIPG) is a pediatric lethal high-grade brainstem glioma with no effective therapies. OLIG2 (oligodendrocyte transcription factor 2) was reported to be critical for the growth of a DIPG cell line CCHMC-DIPG-1. Surprisingly, we found that the CCHMC-DIPG-1 cells express little OLIG2 and exhibit a mesenchymal phenotype, which raised a question regarding the role of OLIG2 in the growth of DIPG cells. METHODS: We evaluated the function of OLIG2 in different DIPG cell lines through molecular and genetic approaches and performed transcriptomic and genomic landscape profiling including whole-genome bisulfite sequencing, RNA-seq, ATAC-seq, and ChIP-seq. shRNA-mediated knockdown and CRISPR-Cas9-mediated knockout approaches were utilized to assess OLIG2 functions in DIPG cell growth. RESULTS: We found that DIPG cells are phenotypically heterogeneous and exhibit the characteristics of distinct malignant gliomas including proneural, classical, and mesenchymal subtypes. OLIG2 knockdown did not impact the growth of CCHMC-DIPG-1 cells, wherein OLIG2 is epigenetically silenced. Moreover, OLIG2 deletion did not substantially impair OLIG2-expressing proneural-like DIPG growth but led to an upregulation of HIPPO-YAP1 and epidermal growth factor receptor (EGFR) signaling and a tumor phenotype shift. Targeting HIPPO-YAP1 and EGFR signaling in OLIG2-deficient DIPG cells inhibited tumor cell growth. CONCLUSIONS: Our data indicate that OLIG2 is dispensable for DIPG growth but regulates the phenotypic switch of DIPG tumor cells. OLIG2 downregulation leads to deregulation of adaptive YAP1 and EGFR signaling. Targeting YAP1 and EGFR pathways inhibits the growth of OLIG2-deficient DIPG cells, pointing to a therapeutic potential by targeting adaptive signaling to treat DIPG tumors with nominal OLIG2 expression.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Brain Stem Neoplasms/genetics , Cell Line , Cell Line, Tumor , Child , Humans , Oligodendrocyte Transcription Factor 2 , Phenotype
7.
Cereb Cortex ; 30(11): 5702-5716, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32564090

ABSTRACT

Axon regeneration is limited in the central nervous system, which hinders the reconstruction of functional circuits following spinal cord injury (SCI). Although various extrinsic molecules to repel axons following SCI have been identified, the role of semaphorins, a major class of axon guidance molecules, has not been thoroughly explored. Here we show that expression of semaphorins, including Sema5a and Sema6d, is elevated after SCI, and genetic deletion of either molecule or their receptors (neuropilin1 and plexinA1, respectively) suppresses axon retraction or dieback in injured corticospinal neurons. We further show that Olig2+ cells are essential for SCI-induced semaphorin expression, and that Olig2 binds to putative enhancer regions of the semaphorin genes. Finally, conditional deletion of Olig2 in the spinal cord reduces the expression of semaphorins, alleviating the axon retraction. These results demonstrate that semaphorins function as axon repellents following SCI, and reveal a novel transcriptional mechanism for controlling semaphorin levels around injured neurons to create zones hostile to axon regrowth.


Subject(s)
Gene Expression Regulation/physiology , Nerve Regeneration/physiology , Oligodendrocyte Transcription Factor 2/metabolism , Semaphorins/biosynthesis , Spinal Cord Injuries/metabolism , Animals , Axons/pathology , Mice , Mice, Inbred C57BL , Pyramidal Tracts/injuries , Pyramidal Tracts/metabolism , Spinal Cord Injuries/pathology
8.
Glia ; 68(8): 1604-1618, 2020 08.
Article in English | MEDLINE | ID: mdl-32460418

ABSTRACT

Oligodendrocytes, the myelinating cells in the vertebrate central nervous system, produce myelin sheaths to enable saltatory propagation of action potentials. The process of oligodendrocyte myelination entails a stepwise progression from precursor specification to differentiation, which is coordinated by a series of transcriptional and chromatin remodeling events. ATP-dependent chromatin remodeling enzymes, which utilize ATP as an energy source to control chromatin dynamics and regulate the accessibility of chromatin to transcriptional regulators, are critical for oligodendrocyte lineage development and regeneration. In this review, we focus on the latest insights into the spatial and temporal specificity of chromatin remodelers during oligodendrocyte development, myelinogenesis, and regeneration. We will also bring together various plausible mechanisms by which lineage specific transcriptional regulators coordinate with chromatin remodeling factors for programming genomic landscapes to specifically modulate these different processes during developmental myelination and remyelination upon injury.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Myelin Sheath/physiology , Oligodendroglia/cytology , Remyelination/physiology , Animals , Cell Differentiation/physiology , Central Nervous System/metabolism , Humans
9.
Cell Chem Biol ; 25(6): 775-786.e5, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29706593

ABSTRACT

Identification of additional uses for existing drugs is a hot topic in drug discovery and a viable alternative to de novo drug development. HAMI3379 is known as an antagonist of the cysteinyl-leukotriene CysLT2 receptor, and was initially developed to treat cardiovascular and inflammatory disorders. In our study we identified HAMI3379 as an antagonist of the orphan G protein-coupled receptor GPR17. HAMI3379 inhibits signaling of recombinant human, rat, and mouse GPR17 across various cellular backgrounds, and of endogenous GPR17 in primary rodent oligodendrocytes. GPR17 blockade by HAMI3379 enhanced maturation of primary rat and mouse oligodendrocytes, but was without effect in oligodendrocytes from GPR17 knockout mice. In human oligodendrocytes prepared from inducible pluripotent stem cells, GPR17 is expressed and its activation impaired oligodendrocyte differentiation. HAMI3379, conversely, efficiently favored human oligodendrocyte differentiation. We propose that HAMI3379 holds promise for pharmacological exploitation of orphan GPR17 to enhance regenerative strategies for the promotion of remyelination in patients.


Subject(s)
Cell Differentiation/drug effects , Cyclohexanecarboxylic Acids/pharmacology , Drug Repositioning , Oligodendroglia/cytology , Oligodendroglia/drug effects , Phthalic Acids/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Cyclohexanecarboxylic Acids/chemistry , Dose-Response Relationship, Drug , Humans , Indoles/chemistry , Indoles/pharmacology , Mice , Mice, Knockout , Molecular Structure , Phthalic Acids/chemistry , Propionates/chemistry , Propionates/pharmacology , Rats , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
10.
Cell Death Dis ; 9(3): 398, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540666

ABSTRACT

Histone deacetylase 3 (HDAC3) plays pivotal roles in cell cycle regulation and is often aberrantly expressed in various cancers including hepatocellular carcinoma (HCC), but little is known about its role in liver regeneration and liver cancer cells proliferation. Using an inducible hepatocyte-selective HDAC3 knockout mouse, we find that lack of HDAC3 dramatically impaired liver regeneration and blocked hepatocyte proliferation in the G1 phase entry. HDAC3 inactivation robustly disrupted the signal transducer and activator of transcription 3 (STAT3) cascade. HDAC3 silencing impaired the ac-STAT3-to-p-STAT3 transition in the cytoplasm, leading to the subsequent breakdown of STAT3 signaling. Furthermore, overexpressed HDAC3 was further associated with increased tumor growth and a poor prognosis in HCC patients. Inhibition of HDAC3 expression reduced liver cancer cells growth and inhibited xenograft tumor growth. Our results suggest that HDAC3 is an important regulator of STAT3-dependent cell proliferation in liver regeneration and cancer. These findings provide novel insights into the HDAC3-STAT3 pathway in liver pathophysiological processes.


Subject(s)
Histone Deacetylases/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/physiopathology , STAT3 Transcription Factor/metabolism , Animals , Cell Proliferation , Hepatocytes/cytology , Hepatocytes/metabolism , Histone Deacetylases/genetics , Humans , Liver Neoplasms/genetics , Liver Regeneration , Male , Mice , Mice, Knockout , STAT3 Transcription Factor/genetics , Signal Transduction
11.
FEBS Lett ; 592(7): 1063-1078, 2018 04.
Article in English | MEDLINE | ID: mdl-29427507

ABSTRACT

Myelination by oligodendrocytes in the central nervous system permits high-fidelity saltatory conduction from neuronal cell bodies to axon terminals. Dysmyelinating and demyelinating disorders impair normal nervous system functions. Consequently, an understanding of oligodendrocyte differentiation that moves beyond the genetic code into the field of epigenetics is essential. Chromatin reprogramming is critical for steering stage-specific differentiation processes during oligodendrocyte development. Fine temporal control of chromatin remodeling through ATP-dependent chromatin remodelers and sequential histone modifiers shapes a chromatin regulatory landscape conducive to oligodendrocyte fate specification, lineage differentiation, and maintenance of cell identity. In this Review, we will focus on the biological functions of ATP-dependent chromatin remodelers and histone deacetylases in myelinating oligodendrocyte development and implications for myelin regeneration in neurodegenerative diseases.


Subject(s)
Chromatin Assembly and Disassembly , Epigenesis, Genetic , Myelin Sheath/metabolism , Neurodegenerative Diseases/metabolism , Oligodendroglia/metabolism , Remyelination , Animals , Cell Differentiation , Humans , Myelin Sheath/pathology , Neurodegenerative Diseases/pathology , Oligodendroglia/pathology
12.
Proc Natl Acad Sci U S A ; 114(41): 10918-10923, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28973878

ABSTRACT

Angiogenesis and vascular remodeling are essential for the establishment of vascular networks during organogenesis. Here we show that the Hippo signaling pathway effectors YAP and TAZ are required, in a gene dosage-dependent manner, for the proliferation and migration of vascular endothelial cells (ECs) during retinal angiogenesis. Intriguingly, nuclear translocation of YAP and TAZ induced by Lats1/2-deletion blocked endothelial migration and phenocopied Yap/Taz-deficient mutants. Furthermore, overexpression of a cytoplasmic form of YAP (YAPS127D) partially rescued the migration defects caused by loss of YAP and TAZ function. Finally, we found that cytoplasmic YAP positively regulated the activity of the small GTPase CDC42, deletion of which caused severe defects in endothelial migration. These findings uncover a previously unrecognized role of cytoplasmic YAP/TAZ in promoting cell migration by activating CDC42 and provide insight into how Hippo signaling in ECs regulates angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Movement , Endothelium, Vascular/cytology , Neovascularization, Physiologic , Phosphoproteins/physiology , Transcription Factors/physiology , cdc42 GTP-Binding Protein/physiology , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Proliferation , Endothelium, Vascular/physiology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Phosphoproteins/genetics , Phosphoproteins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins
13.
Bio Protoc ; 7(14): e2404, 2017 Jul 20.
Article in English | MEDLINE | ID: mdl-34541135

ABSTRACT

Glioblastoma (GBM) is the most common primary brain cancer in adults and has a poor prognosis. It is characterized by a high degree of cellular infiltration that leads to tumor recurrence, atypical hyperplasia, necrosis, and angiogenesis. Despite aggressive treatment modalities, current therapies are ineffective for GBM. Mouse GBM models not only provide a better understanding in the mechanisms of gliomagenesis, but also facilitate the drug discovery for treating this deadly cancer. A retroviral vector system that expresses PDGFBB (Platelet-derived growth factor BB) and inactivates PTEN (Phosphatase and tensin homolog) and P53 tumor suppressors provides a rapid and efficient induction of glioma in mice with full penetrance. In this protocol, we describe a simple and practical method for inducing GBM formation by retrovirus injection in the murine brain. This system gives a spatial and temporal control over the induction of glioma and allows the assessment of therapeutic effects with a bioluminescent reporter.

14.
J Neurosci ; 34(25): 8467-73, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24948802

ABSTRACT

Oligodendrocytes are myelin-forming glia that ensheath the axons of neurons in the CNS. Recent studies have revealed that Wnt/ß-catenin signaling plays important roles in oligodendrocyte development and myelin formation. However, there are conflicting reports on the specific function of Wnt signaling components in oligodendrocyte specification and differentiation. In the present study, we demonstrate that activation of ß-catenin in neural progenitor cells before gliogenesis inhibits the generation of oligodendrocyte progenitors (OLPs) in mice. Once OLPs are formed, ß-catenin becomes necessary for oligodendrocyte differentiation. Disruption of ß-catenin signaling instead leads to a significant delay of oligodendrocyte maturation. These findings suggest that Wnt/ß-catenin pathway regulates oligodendrocyte development in a stage-dependent manner.


Subject(s)
Cell Differentiation/physiology , Oligodendroglia/physiology , Signal Transduction/physiology , Wnt Proteins/physiology , beta Catenin/physiology , Animals , Cell Movement/physiology , Cells, Cultured , Female , Male , Mice , Mice, Transgenic , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/physiology
15.
J Neurosci Methods ; 178(1): 87-98, 2009 Mar 30.
Article in English | MEDLINE | ID: mdl-19100769

ABSTRACT

Galactocerebroside (GalC) and its sulfated derivative sulfatide (SUL) are galactosphingolipids abundantly expressed in oligodendrocytes (OLs). Despite their biological importance in OL development and function, attempts to visualize GalC/SUL in tissue sections have met with limited success. This is at least in part because permeabilization of tissue sections with detergents such as Triton X-100 results in significant degradation of GalC/SUL immunoreactivity. Here we establish a novel method that enables visualization of endogenous GalC/SUL in OLs and myelin throughout the entire depth of brain sections. We show that treating brain sections with the cholesterol-specific detergent digitonin instead of Triton X-100 or methanol leads to efficient antibody penetration into tissue sections without disrupting GalC/SUL immunoreactivity. We also determine the optimal concentrations of digitonin using confocal microscopy. With our method, the morphology and the number of GalC/SUL-expressing OLs can be visualized three-dimensionally. Furthermore, our method is applicable to double immunostaining with anti-GalC/SUL antibody and other antibodies which recognize intracellular antigens. Our simple method using digitonin should prove to be useful in enabling detailed examination of GalC/SUL expression in the brain in both physiological and pathological conditions.


Subject(s)
Brain/cytology , Detergents/pharmacology , Digitonin/pharmacology , Galactosylceramides/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Animals , Animals, Newborn , Autophagy-Related Proteins , Dose-Response Relationship, Drug , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Myelin Basic Protein/metabolism , O Antigens/metabolism , Octoxynol/pharmacology , Phosphopyruvate Hydratase/metabolism , Sulfoglycosphingolipids/metabolism
16.
Mol Cell Neurosci ; 39(1): 40-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18588982

ABSTRACT

Termination of developmental plasticity occurs at specific points in development, and the mechanisms responsible for it are not well understood. One hypothesis that has been proposed is that oligodendrocytes (OLs) play an important role. Consistent with this, we found that OLs appeared in the mouse somatosensory cortex at the end of the critical period for whisker lesion-induced barrel structural plasticity. To test this hypothesis, we used two mouse lines with defective OL differentiation: Olig1-deficient and jimpy. In Olig1-deficient mice, although OLs were totally absent, the termination of lesion-induced plasticity was not delayed. The timing was normal even when the cytoarchitectonic barrel formation was temporarily blocked by pharmacological treatment in Olig1-deficient mice. Furthermore, the termination was not delayed in jimpy mice. These results demonstrate that, even though OLs appear at the end of the critical period, OLs are not intrinsically necessary for the termination of lesion-induced plasticity. Our findings underscore a mechanistic distinction between the termination of thalamocortical axonal plasticity in the barrel cortex and that in the visual cortex, in which OL-derived Nogo-A/B was recently suggested to be essential.


Subject(s)
Neuronal Plasticity/physiology , Oligodendroglia/metabolism , Somatosensory Cortex , Vibrissae/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Clorgyline/metabolism , Mice , Mice, Inbred C57BL , Mice, Jimpy , Mice, Knockout , Monoamine Oxidase Inhibitors/metabolism , Oligodendroglia/cytology , Somatosensory Cortex/cytology , Somatosensory Cortex/pathology , Somatosensory Cortex/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...