Cargando…

AhR Deletion Promotes Aberrant Morphogenesis and Synaptic Activity of Adult-Generated Granule Neurons and Impairs Hippocampus-Dependent Memory

Newborn granule cells are continuously produced in the subgranular zone of dentate gyrus throughout life. Once these cells mature, they integrate into pre-existing circuits modulating hippocampus-dependent memory. Subsequently, mechanisms controlling generation and maturation of newborn cells are es...

Descripción completa

Detalles Bibliográficos
Autores principales: de la Parra, Juan, Cuartero, María I., Pérez-Ruiz, Alberto, García-Culebras, Alicia, Martín, Ricardo, Sánchez-Prieto, José, García-Segura, Juan M., Lizasoain, Ignacio, Moro, María A.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Society for Neuroscience 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6140122/
https://www.ncbi.nlm.nih.gov/pubmed/30225360
http://dx.doi.org/10.1523/ENEURO.0370-17.2018
_version_ 1783355561512796160
author de la Parra, Juan
Cuartero, María I.
Pérez-Ruiz, Alberto
García-Culebras, Alicia
Martín, Ricardo
Sánchez-Prieto, José
García-Segura, Juan M.
Lizasoain, Ignacio
Moro, María A.
author_facet de la Parra, Juan
Cuartero, María I.
Pérez-Ruiz, Alberto
García-Culebras, Alicia
Martín, Ricardo
Sánchez-Prieto, José
García-Segura, Juan M.
Lizasoain, Ignacio
Moro, María A.
author_sort de la Parra, Juan
collection PubMed
description Newborn granule cells are continuously produced in the subgranular zone of dentate gyrus throughout life. Once these cells mature, they integrate into pre-existing circuits modulating hippocampus-dependent memory. Subsequently, mechanisms controlling generation and maturation of newborn cells are essential for proper hippocampal function. Therefore, we have studied the role of aryl hydrocarbon receptor (AhR), a ligand-activated bHLH-PAS transcription factor, in hippocampus-dependent memory and granule neuronal morphology and function using genetic loss-of-function approaches based on constitutive and inducible-nestin AhR(–/–) mice. The results presented here show that the impaired hippocampus-dependent memory in AhR absence is not due to its effects on neurogenesis but to aberrant dendritic arborization and an increased spine density, albeit with a lower number of mature mushrooms spines in newborn granule cells, a finding that is associated with an immature electrophysiological phenotype. Together, our data strongly suggest that AhR plays a pivotal role in the regulation of hippocampal function, by controlling hippocampal granule neuron morphology and synaptic maturation.
format Online
Article
Text
id pubmed-6140122
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Society for Neuroscience
record_format MEDLINE/PubMed
spelling pubmed-61401222018-09-17 AhR Deletion Promotes Aberrant Morphogenesis and Synaptic Activity of Adult-Generated Granule Neurons and Impairs Hippocampus-Dependent Memory de la Parra, Juan Cuartero, María I. Pérez-Ruiz, Alberto García-Culebras, Alicia Martín, Ricardo Sánchez-Prieto, José García-Segura, Juan M. Lizasoain, Ignacio Moro, María A. eNeuro New Research Newborn granule cells are continuously produced in the subgranular zone of dentate gyrus throughout life. Once these cells mature, they integrate into pre-existing circuits modulating hippocampus-dependent memory. Subsequently, mechanisms controlling generation and maturation of newborn cells are essential for proper hippocampal function. Therefore, we have studied the role of aryl hydrocarbon receptor (AhR), a ligand-activated bHLH-PAS transcription factor, in hippocampus-dependent memory and granule neuronal morphology and function using genetic loss-of-function approaches based on constitutive and inducible-nestin AhR(–/–) mice. The results presented here show that the impaired hippocampus-dependent memory in AhR absence is not due to its effects on neurogenesis but to aberrant dendritic arborization and an increased spine density, albeit with a lower number of mature mushrooms spines in newborn granule cells, a finding that is associated with an immature electrophysiological phenotype. Together, our data strongly suggest that AhR plays a pivotal role in the regulation of hippocampal function, by controlling hippocampal granule neuron morphology and synaptic maturation. Society for Neuroscience 2018-08-22 /pmc/articles/PMC6140122/ /pubmed/30225360 http://dx.doi.org/10.1523/ENEURO.0370-17.2018 Text en Copyright © 2018 de la Parra et al. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license (http://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.
spellingShingle New Research
de la Parra, Juan
Cuartero, María I.
Pérez-Ruiz, Alberto
García-Culebras, Alicia
Martín, Ricardo
Sánchez-Prieto, José
García-Segura, Juan M.
Lizasoain, Ignacio
Moro, María A.
AhR Deletion Promotes Aberrant Morphogenesis and Synaptic Activity of Adult-Generated Granule Neurons and Impairs Hippocampus-Dependent Memory
title AhR Deletion Promotes Aberrant Morphogenesis and Synaptic Activity of Adult-Generated Granule Neurons and Impairs Hippocampus-Dependent Memory
title_full AhR Deletion Promotes Aberrant Morphogenesis and Synaptic Activity of Adult-Generated Granule Neurons and Impairs Hippocampus-Dependent Memory
title_fullStr AhR Deletion Promotes Aberrant Morphogenesis and Synaptic Activity of Adult-Generated Granule Neurons and Impairs Hippocampus-Dependent Memory
title_full_unstemmed AhR Deletion Promotes Aberrant Morphogenesis and Synaptic Activity of Adult-Generated Granule Neurons and Impairs Hippocampus-Dependent Memory
title_short AhR Deletion Promotes Aberrant Morphogenesis and Synaptic Activity of Adult-Generated Granule Neurons and Impairs Hippocampus-Dependent Memory
title_sort ahr deletion promotes aberrant morphogenesis and synaptic activity of adult-generated granule neurons and impairs hippocampus-dependent memory
topic New Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6140122/
https://www.ncbi.nlm.nih.gov/pubmed/30225360
http://dx.doi.org/10.1523/ENEURO.0370-17.2018
work_keys_str_mv AT delaparrajuan ahrdeletionpromotesaberrantmorphogenesisandsynapticactivityofadultgeneratedgranuleneuronsandimpairshippocampusdependentmemory
AT cuarteromariai ahrdeletionpromotesaberrantmorphogenesisandsynapticactivityofadultgeneratedgranuleneuronsandimpairshippocampusdependentmemory
AT perezruizalberto ahrdeletionpromotesaberrantmorphogenesisandsynapticactivityofadultgeneratedgranuleneuronsandimpairshippocampusdependentmemory
AT garciaculebrasalicia ahrdeletionpromotesaberrantmorphogenesisandsynapticactivityofadultgeneratedgranuleneuronsandimpairshippocampusdependentmemory
AT martinricardo ahrdeletionpromotesaberrantmorphogenesisandsynapticactivityofadultgeneratedgranuleneuronsandimpairshippocampusdependentmemory
AT sanchezprietojose ahrdeletionpromotesaberrantmorphogenesisandsynapticactivityofadultgeneratedgranuleneuronsandimpairshippocampusdependentmemory
AT garciasegurajuanm ahrdeletionpromotesaberrantmorphogenesisandsynapticactivityofadultgeneratedgranuleneuronsandimpairshippocampusdependentmemory
AT lizasoainignacio ahrdeletionpromotesaberrantmorphogenesisandsynapticactivityofadultgeneratedgranuleneuronsandimpairshippocampusdependentmemory
AT moromariaa ahrdeletionpromotesaberrantmorphogenesisandsynapticactivityofadultgeneratedgranuleneuronsandimpairshippocampusdependentmemory