Cargando…

Estradiol Attenuates Ischemia-Induced Death of Hippocampal Neurons and Enhances Synaptic Transmission in Aged, Long-Term Hormone-Deprived Female Rats

BACKGROUND: Transient global forebrain ischemia causes selective, delayed death of hippocampal CA1 pyramidal neurons, and the ovarian hormone 17β-estradiol (E2) reduces neuronal loss in young and middle-aged females. The neuroprotective efficacy of E2 after a prolonged period of hormone deprivation...

Descripción completa

Detalles Bibliográficos
Autores principales: Inagaki, Tomoko, Kaneko, Naoki, Zukin, R. Suzanne, Castillo, Pablo E., Etgen, Anne M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3366987/
https://www.ncbi.nlm.nih.gov/pubmed/22675505
http://dx.doi.org/10.1371/journal.pone.0038018
_version_ 1782234802449022976
author Inagaki, Tomoko
Kaneko, Naoki
Zukin, R. Suzanne
Castillo, Pablo E.
Etgen, Anne M.
author_facet Inagaki, Tomoko
Kaneko, Naoki
Zukin, R. Suzanne
Castillo, Pablo E.
Etgen, Anne M.
author_sort Inagaki, Tomoko
collection PubMed
description BACKGROUND: Transient global forebrain ischemia causes selective, delayed death of hippocampal CA1 pyramidal neurons, and the ovarian hormone 17β-estradiol (E2) reduces neuronal loss in young and middle-aged females. The neuroprotective efficacy of E2 after a prolonged period of hormone deprivation is controversial, and few studies examine this issue in aged animals given E2 treatment after induction of ischemia. METHODOLOGY/PRINCIPAL FINDINGS: The present study investigated the neuroprotective effects of E2 administered immediately after global ischemia in aged female rats (15–18 months) after 6 months of hormone deprivation. We also used electrophysiological methods to assess whether CA1 synapses in the aging hippocampus remain responsive to E2 after prolonged hormone withdrawal. Animals were ovariohysterectomized and underwent 10 min global ischemia 6 months later. A single dose of E2 (2.25 µg) infused intraventricularly after reperfusion significantly increased cell survival, with 45% of CA1 neurons surviving vs 15% in controls. Ischemia also induced moderate loss of CA3/CA4 pyramidal cells. Bath application of 1 nM E2 onto brain slices derived from non-ischemic aged females after 6 months of hormone withdrawal significantly enhanced excitatory transmission at CA1 synapses evoked by Schaffer collateral stimulation, and normal long-term potentiation (LTP) was induced. The magnitude of LTP and of E2 enhancement of field excitatory postsynaptic potentials was indistinguishable from that recorded in slices from young rats. CONCLUSIONS/SIGNIFICANCE: The data demonstrate that 1) acute post-ischemic infusion of E2 into the brain ventricles is neuroprotective in aged rats after 6 months of hormone deprivation; and 2) E2 enhances synaptic transmission in CA1 pyramidal neurons of aged long-term hormone deprived females. These findings provide evidence that the aging hippocampus remains responsive to E2 administered either in vivo or in vitro even after prolonged periods of hormone withdrawal.
format Online
Article
Text
id pubmed-3366987
institution National Center for Biotechnology Information
language English
publishDate 2012
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-33669872012-06-06 Estradiol Attenuates Ischemia-Induced Death of Hippocampal Neurons and Enhances Synaptic Transmission in Aged, Long-Term Hormone-Deprived Female Rats Inagaki, Tomoko Kaneko, Naoki Zukin, R. Suzanne Castillo, Pablo E. Etgen, Anne M. PLoS One Research Article BACKGROUND: Transient global forebrain ischemia causes selective, delayed death of hippocampal CA1 pyramidal neurons, and the ovarian hormone 17β-estradiol (E2) reduces neuronal loss in young and middle-aged females. The neuroprotective efficacy of E2 after a prolonged period of hormone deprivation is controversial, and few studies examine this issue in aged animals given E2 treatment after induction of ischemia. METHODOLOGY/PRINCIPAL FINDINGS: The present study investigated the neuroprotective effects of E2 administered immediately after global ischemia in aged female rats (15–18 months) after 6 months of hormone deprivation. We also used electrophysiological methods to assess whether CA1 synapses in the aging hippocampus remain responsive to E2 after prolonged hormone withdrawal. Animals were ovariohysterectomized and underwent 10 min global ischemia 6 months later. A single dose of E2 (2.25 µg) infused intraventricularly after reperfusion significantly increased cell survival, with 45% of CA1 neurons surviving vs 15% in controls. Ischemia also induced moderate loss of CA3/CA4 pyramidal cells. Bath application of 1 nM E2 onto brain slices derived from non-ischemic aged females after 6 months of hormone withdrawal significantly enhanced excitatory transmission at CA1 synapses evoked by Schaffer collateral stimulation, and normal long-term potentiation (LTP) was induced. The magnitude of LTP and of E2 enhancement of field excitatory postsynaptic potentials was indistinguishable from that recorded in slices from young rats. CONCLUSIONS/SIGNIFICANCE: The data demonstrate that 1) acute post-ischemic infusion of E2 into the brain ventricles is neuroprotective in aged rats after 6 months of hormone deprivation; and 2) E2 enhances synaptic transmission in CA1 pyramidal neurons of aged long-term hormone deprived females. These findings provide evidence that the aging hippocampus remains responsive to E2 administered either in vivo or in vitro even after prolonged periods of hormone withdrawal. Public Library of Science 2012-06-04 /pmc/articles/PMC3366987/ /pubmed/22675505 http://dx.doi.org/10.1371/journal.pone.0038018 Text en Inagaki et al. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Inagaki, Tomoko
Kaneko, Naoki
Zukin, R. Suzanne
Castillo, Pablo E.
Etgen, Anne M.
Estradiol Attenuates Ischemia-Induced Death of Hippocampal Neurons and Enhances Synaptic Transmission in Aged, Long-Term Hormone-Deprived Female Rats
title Estradiol Attenuates Ischemia-Induced Death of Hippocampal Neurons and Enhances Synaptic Transmission in Aged, Long-Term Hormone-Deprived Female Rats
title_full Estradiol Attenuates Ischemia-Induced Death of Hippocampal Neurons and Enhances Synaptic Transmission in Aged, Long-Term Hormone-Deprived Female Rats
title_fullStr Estradiol Attenuates Ischemia-Induced Death of Hippocampal Neurons and Enhances Synaptic Transmission in Aged, Long-Term Hormone-Deprived Female Rats
title_full_unstemmed Estradiol Attenuates Ischemia-Induced Death of Hippocampal Neurons and Enhances Synaptic Transmission in Aged, Long-Term Hormone-Deprived Female Rats
title_short Estradiol Attenuates Ischemia-Induced Death of Hippocampal Neurons and Enhances Synaptic Transmission in Aged, Long-Term Hormone-Deprived Female Rats
title_sort estradiol attenuates ischemia-induced death of hippocampal neurons and enhances synaptic transmission in aged, long-term hormone-deprived female rats
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3366987/
https://www.ncbi.nlm.nih.gov/pubmed/22675505
http://dx.doi.org/10.1371/journal.pone.0038018
work_keys_str_mv AT inagakitomoko estradiolattenuatesischemiainduceddeathofhippocampalneuronsandenhancessynaptictransmissioninagedlongtermhormonedeprivedfemalerats
AT kanekonaoki estradiolattenuatesischemiainduceddeathofhippocampalneuronsandenhancessynaptictransmissioninagedlongtermhormonedeprivedfemalerats
AT zukinrsuzanne estradiolattenuatesischemiainduceddeathofhippocampalneuronsandenhancessynaptictransmissioninagedlongtermhormonedeprivedfemalerats
AT castillopabloe estradiolattenuatesischemiainduceddeathofhippocampalneuronsandenhancessynaptictransmissioninagedlongtermhormonedeprivedfemalerats
AT etgenannem estradiolattenuatesischemiainduceddeathofhippocampalneuronsandenhancessynaptictransmissioninagedlongtermhormonedeprivedfemalerats