Neuronal cell depolarization induces intragenic chromatin modifications affecting NCAM alternative splicing

In search for physiological pathways affecting alternative splicing through its kinetic coupling with transcription, we found that membrane depolarization of neuronal cells triggers the skipping of exon 18 from the neural cell adhesion molecule (NCAM) mRNA, independently of the calcium/calmodulin pr...

Descripción completa

Guardado en:
Detalles Bibliográficos
Publicado: 2009
Materias:
rat
Acceso en línea:https://bibliotecadigital.exactas.uba.ar/collection/paper/document/paper_00278424_v106_n11_p4325_Schor
http://hdl.handle.net/20.500.12110/paper_00278424_v106_n11_p4325_Schor
Aporte de:
id paper:paper_00278424_v106_n11_p4325_Schor
record_format dspace
spelling paper:paper_00278424_v106_n11_p4325_Schor2023-06-08T14:54:22Z Neuronal cell depolarization induces intragenic chromatin modifications affecting NCAM alternative splicing Histone acetylation MRNA processing Neuronal excitation Transcription calcium calmodulin dependent protein kinase messenger RNA nerve cell adhesion molecule RNA polymerase II trichostatin A acetylation alternative RNA splicing animal cell article cell differentiation cell elongation cell function cell membrane depolarization chromatin chromatin structure controlled study embryo exon gene location genetic epigenesis methylation mutant nerve cell nonhuman priority journal promoter region protein folding rat RNA splicing transcription initiation Acetylation Alternative Splicing Animals Chromatin Epigenesis, Genetic Exons Histones Membrane Potentials Neural Cell Adhesion Molecules Neurons Rats RNA Polymerase II In search for physiological pathways affecting alternative splicing through its kinetic coupling with transcription, we found that membrane depolarization of neuronal cells triggers the skipping of exon 18 from the neural cell adhesion molecule (NCAM) mRNA, independently of the calcium/calmodulin protein kinase IV pathway. We show that this exon responds to RNA polymerase II elongation, because its inclusion is increased by a slow polymerase II mutant. Depolarization affects the chromatin template in a specific way, by causing H3K9 hyper-acetylation restricted to an internal region of the NCAM gene surrounding the alternative exon. This intragenic histone hyper-acetylation is not paralleled by acetylation at the promoter, is associated with chromatin relaxation, and is linked to H3K36 trimethylation. The effects on acetylation and splicing fully revert when the depolarizing conditions are withdrawn and can be both duplicated and potentiated by the histone deacetylase inhibitor trichostatin A. Our results are consistent with a mechanism involving the kinetic coupling of splicing and transcription in response to depolarization through intragenic epigenetic changes on a gene that is relevant for the differentiation and function of neuronal cells. 2009 https://bibliotecadigital.exactas.uba.ar/collection/paper/document/paper_00278424_v106_n11_p4325_Schor http://hdl.handle.net/20.500.12110/paper_00278424_v106_n11_p4325_Schor
institution Universidad de Buenos Aires
institution_str I-28
repository_str R-134
collection Biblioteca Digital - Facultad de Ciencias Exactas y Naturales (UBA)
topic Histone acetylation
MRNA processing
Neuronal excitation
Transcription
calcium calmodulin dependent protein kinase
messenger RNA
nerve cell adhesion molecule
RNA polymerase II
trichostatin A
acetylation
alternative RNA splicing
animal cell
article
cell differentiation
cell elongation
cell function
cell membrane depolarization
chromatin
chromatin structure
controlled study
embryo
exon
gene location
genetic epigenesis
methylation
mutant
nerve cell
nonhuman
priority journal
promoter region
protein folding
rat
RNA splicing
transcription initiation
Acetylation
Alternative Splicing
Animals
Chromatin
Epigenesis, Genetic
Exons
Histones
Membrane Potentials
Neural Cell Adhesion Molecules
Neurons
Rats
RNA Polymerase II
spellingShingle Histone acetylation
MRNA processing
Neuronal excitation
Transcription
calcium calmodulin dependent protein kinase
messenger RNA
nerve cell adhesion molecule
RNA polymerase II
trichostatin A
acetylation
alternative RNA splicing
animal cell
article
cell differentiation
cell elongation
cell function
cell membrane depolarization
chromatin
chromatin structure
controlled study
embryo
exon
gene location
genetic epigenesis
methylation
mutant
nerve cell
nonhuman
priority journal
promoter region
protein folding
rat
RNA splicing
transcription initiation
Acetylation
Alternative Splicing
Animals
Chromatin
Epigenesis, Genetic
Exons
Histones
Membrane Potentials
Neural Cell Adhesion Molecules
Neurons
Rats
RNA Polymerase II
Neuronal cell depolarization induces intragenic chromatin modifications affecting NCAM alternative splicing
topic_facet Histone acetylation
MRNA processing
Neuronal excitation
Transcription
calcium calmodulin dependent protein kinase
messenger RNA
nerve cell adhesion molecule
RNA polymerase II
trichostatin A
acetylation
alternative RNA splicing
animal cell
article
cell differentiation
cell elongation
cell function
cell membrane depolarization
chromatin
chromatin structure
controlled study
embryo
exon
gene location
genetic epigenesis
methylation
mutant
nerve cell
nonhuman
priority journal
promoter region
protein folding
rat
RNA splicing
transcription initiation
Acetylation
Alternative Splicing
Animals
Chromatin
Epigenesis, Genetic
Exons
Histones
Membrane Potentials
Neural Cell Adhesion Molecules
Neurons
Rats
RNA Polymerase II
description In search for physiological pathways affecting alternative splicing through its kinetic coupling with transcription, we found that membrane depolarization of neuronal cells triggers the skipping of exon 18 from the neural cell adhesion molecule (NCAM) mRNA, independently of the calcium/calmodulin protein kinase IV pathway. We show that this exon responds to RNA polymerase II elongation, because its inclusion is increased by a slow polymerase II mutant. Depolarization affects the chromatin template in a specific way, by causing H3K9 hyper-acetylation restricted to an internal region of the NCAM gene surrounding the alternative exon. This intragenic histone hyper-acetylation is not paralleled by acetylation at the promoter, is associated with chromatin relaxation, and is linked to H3K36 trimethylation. The effects on acetylation and splicing fully revert when the depolarizing conditions are withdrawn and can be both duplicated and potentiated by the histone deacetylase inhibitor trichostatin A. Our results are consistent with a mechanism involving the kinetic coupling of splicing and transcription in response to depolarization through intragenic epigenetic changes on a gene that is relevant for the differentiation and function of neuronal cells.
title Neuronal cell depolarization induces intragenic chromatin modifications affecting NCAM alternative splicing
title_short Neuronal cell depolarization induces intragenic chromatin modifications affecting NCAM alternative splicing
title_full Neuronal cell depolarization induces intragenic chromatin modifications affecting NCAM alternative splicing
title_fullStr Neuronal cell depolarization induces intragenic chromatin modifications affecting NCAM alternative splicing
title_full_unstemmed Neuronal cell depolarization induces intragenic chromatin modifications affecting NCAM alternative splicing
title_sort neuronal cell depolarization induces intragenic chromatin modifications affecting ncam alternative splicing
publishDate 2009
url https://bibliotecadigital.exactas.uba.ar/collection/paper/document/paper_00278424_v106_n11_p4325_Schor
http://hdl.handle.net/20.500.12110/paper_00278424_v106_n11_p4325_Schor
_version_ 1768543548051816448