Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 122023 04 24.
Article in English | MEDLINE | ID: mdl-37092728

ABSTRACT

The functional maturation of neurons is a prolonged process that extends past the mitotic exit and is mediated by the chromatin-dependent orchestration of gene transcription programs. We find that expression of this maturation gene program in mouse cerebellar granule neurons (CGNs) requires dynamic changes in the genomic distribution of histone H3 lysine 27 trimethylation (H3K27me3), demonstrating a function for this chromatin modification beyond its role in cell fate specification. The developmental loss of H3K27me3 at promoters of genes activated as CGNs mature is facilitated by the lysine demethylase and ASD-risk gene, Kdm6b. Interestingly, inhibition of the H3K27 methyltransferase EZH2 in newborn CGNs not only blocks the repression of progenitor genes but also impairs the induction of mature CGN genes, showing the importance of bidirectional H3K27me3 regulation across the genome. These data demonstrate that H3K27me3 turnover in developing postmitotic neurons regulates the temporal coordination of gene expression programs that underlie functional neuronal maturation.


Subject(s)
Histones , Lysine , Animals , Mice , Histones/metabolism , Lysine/metabolism , Chromatin , Cell Differentiation/genetics , Neurons/metabolism
2.
J Vis Exp ; (172)2021 06 09.
Article in English | MEDLINE | ID: mdl-34180898

ABSTRACT

Neurons undergo dynamic changes in their structure and function during brain development to form appropriate connections with other cells. The rodent cerebellum is an ideal system to track the development and morphogenesis of a single cell type, the cerebellar granule neuron (CGN), across time. Here, in vivo electroporation of granule neuron progenitors in the developing mouse cerebellum was employed to sparsely label cells for subsequent morphological analyses. The efficacy of this technique is demonstrated in its ability to showcase key developmental stages of CGN maturation, with a specific focus on the formation of dendritic claws, which are specialized structures where these cells receive the majority of their synaptic inputs. In addition to providing snapshots of CGN synaptic structures throughout cerebellar development, this technique can be adapted to genetically manipulate granule neurons in a cell-autonomous manner to study the role of any gene of interest and its effect on CGN morphology, claw development, and synaptogenesis.


Subject(s)
Cerebellum , Neurons , Animals , Cytoplasmic Granules , Electroporation , Mice , Synapses
3.
Elife ; 72018 05 29.
Article in English | MEDLINE | ID: mdl-29809135

ABSTRACT

Sensory experience plays an important role in shaping neural circuitry by affecting the synaptic connectivity and intrinsic properties of individual neurons. Identifying the molecular players responsible for converting external stimuli into altered neuronal output remains a crucial step in understanding experience-dependent plasticity and circuit function. Here, we investigate the role of the activity-regulated, non-canonical Ras-like GTPase Rem2 in visual circuit plasticity. We demonstrate that Rem2-/- mice fail to exhibit normal ocular dominance plasticity during the critical period. At the cellular level, our data establish a cell-autonomous role for Rem2 in regulating intrinsic excitability of layer 2/3 pyramidal neurons, prior to changes in synaptic function. Consistent with these findings, both in vitro and in vivo recordings reveal increased spontaneous firing rates in the absence of Rem2. Taken together, our data demonstrate that Rem2 is a key molecule that regulates neuronal excitability and circuit function in the context of changing sensory experience.


Subject(s)
Monomeric GTP-Binding Proteins/genetics , Nerve Net/metabolism , Neuronal Plasticity/genetics , Pyramidal Cells/metabolism , Sensory Receptor Cells/metabolism , Visual Cortex/metabolism , Action Potentials/physiology , Animals , Female , Gene Expression Regulation , Male , Mice , Mice, Knockout , Monomeric GTP-Binding Proteins/deficiency , Nerve Net/cytology , Primary Cell Culture , Pyramidal Cells/cytology , Rats , Sensory Receptor Cells/cytology , Synapses/genetics , Synapses/metabolism , Visual Cortex/cytology
4.
Trends Neurosci ; 41(5): 311-324, 2018 05.
Article in English | MEDLINE | ID: mdl-29530320

ABSTRACT

Neurons are dynamic cells that respond and adapt to stimuli throughout their long postmitotic lives. The structural and functional plasticity of neurons requires the regulated transcription of new gene products, and dysregulation of transcription in either the developing or adult brain impairs cognition. We discuss how mechanisms of chromatin regulation help to orchestrate the transcriptional programs that underlie the maturation of developing neurons and the plasticity of adult neurons. We review how chromatin regulation acts locally to modulate the expression of specific genes and more broadly to coordinate gene expression programs during transitions between cellular states. These data highlight the importance of epigenetic transcriptional mechanisms in postmitotic neurons. We suggest areas where emerging methods may advance understanding in the future.


Subject(s)
Chromatin/metabolism , Neurogenesis/physiology , Neuronal Plasticity/physiology , Neurons/metabolism , Animals , Humans , Neurogenesis/genetics , Neuronal Plasticity/genetics
5.
Mol Cell Neurosci ; 87: 4-17, 2018 03.
Article in English | MEDLINE | ID: mdl-29254825

ABSTRACT

The histone H3 lysine 27 (H3K27) demethylase Kdm6b (Jmjd3) can promote cellular differentiation, however its physiological functions in neurons remain to be fully determined. We studied the expression and function of Kdm6b in differentiating granule neurons of the developing postnatal mouse cerebellum. At postnatal day 7, Kdm6b is expressed throughout the layers of the developing cerebellar cortex, but its expression is upregulated in newborn cerebellar granule neurons (CGNs). Atoh1-Cre mediated conditional knockout of Kdm6b in CGN precursors either alone or in combination with Kdm6a did not disturb the gross morphological development of the cerebellum. Furthermore, RNAi-mediated knockdown of Kdm6b in cultured CGN precursors did not alter the induced expression of early neuronal marker genes upon cell cycle exit. By contrast, knockdown of Kdm6b significantly impaired the induction of a mature neuronal gene expression program, which includes gene products required for functional synapse maturation. Loss of Kdm6b also impaired the ability of Brain-Derived Neurotrophic Factor (BDNF) to induce expression of Grin2c and Tiam1 in maturing CGNs. Taken together, these data reveal a previously unknown role for Kdm6b in the postmitotic stages of CGN maturation and suggest that Kdm6b may work, at least in part, by a transcriptional mechanism that promotes gene sensitivity to regulation by BDNF.


Subject(s)
Histone Demethylases/genetics , Histones/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Neurons/metabolism , Animals , Cytoplasmic Granules/metabolism , Gene Expression/genetics , Histones/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...