Cargando…

ERα Signaling Is Required for TrkB-Mediated Hippocampal Neuroprotection in Female Neonatal Mice after Hypoxic Ischemic Encephalopathy123

Male neonate brains are more susceptible to the effects of perinatal asphyxia resulting in hypoxia and ischemia (HI)-related brain injury. The relative resistance of female neonatal brains to adverse consequences of HI suggests that there are sex-specific mechanisms that afford females greater neuro...

Descripción completa

Detalles Bibliográficos
Autores principales: Cikla, Ulas, Chanana, Vishal, Kintner, Douglas B., Udho, Eshwar, Eickhoff, Jens, Sun, Wendy, Marquez, Stephanie, Covert, Lucia, Otles, Arel, Shapiro, Robert A., Ferrazzano, Peter, Vemuganti, Raghu, Levine, Jon E., Cengiz, Pelin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Society for Neuroscience 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4731462/
https://www.ncbi.nlm.nih.gov/pubmed/26839918
http://dx.doi.org/10.1523/ENEURO.0025-15.2015
_version_ 1782412545769865216
author Cikla, Ulas
Chanana, Vishal
Kintner, Douglas B.
Udho, Eshwar
Eickhoff, Jens
Sun, Wendy
Marquez, Stephanie
Covert, Lucia
Otles, Arel
Shapiro, Robert A.
Ferrazzano, Peter
Vemuganti, Raghu
Levine, Jon E.
Cengiz, Pelin
author_facet Cikla, Ulas
Chanana, Vishal
Kintner, Douglas B.
Udho, Eshwar
Eickhoff, Jens
Sun, Wendy
Marquez, Stephanie
Covert, Lucia
Otles, Arel
Shapiro, Robert A.
Ferrazzano, Peter
Vemuganti, Raghu
Levine, Jon E.
Cengiz, Pelin
author_sort Cikla, Ulas
collection PubMed
description Male neonate brains are more susceptible to the effects of perinatal asphyxia resulting in hypoxia and ischemia (HI)-related brain injury. The relative resistance of female neonatal brains to adverse consequences of HI suggests that there are sex-specific mechanisms that afford females greater neuroprotection and/or facilitates recovery post-HI. We hypothesized that HI preferentially induces estrogen receptor α (ERα) expression in female neonatal hippocampi and that ERα is coupled to Src family kinase (SFK) activation that in turn augments phosphorylation of the TrkB and thereby results in decreased apoptosis. After inducing the Vannucci’s HI model on P9 (C57BL/6J) mice, female and male ERα wild-type (ERα(+/+)) or ERα null mutant (ERα(−/−)) mice received vehicle control or the selective TrkB agonist 7,8-dihydroxyflavone (7,8-DHF). Hippocampi were collected for analysis of mRNA of ERα and BDNF, protein levels of ERα, p-TrkB, p-src, and cleaved caspase 3 (c-caspase-3) post-HI. Our results demonstrate that: (1) HI differentially induces ERα expression in the hippocampus of the female versus male neonate, (2) src and TrkB phosphorylation post-HI is greater in females than in males after 7,8-DHF therapy, (3) src and TrkB phosphorylation post-HI depend on the presence of ERα, and (4) TrkB agonist therapy decreases the c-caspase-3 only in ERα(+/+) female mice hippocampus. Together, these observations provide evidence that female-specific induction of ERα expression confers neuroprotection with TrkB agonist therapy via SFK activation and account for improved functional outcomes in female neonates post-HI.
format Online
Article
Text
id pubmed-4731462
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Society for Neuroscience
record_format MEDLINE/PubMed
spelling pubmed-47314622016-02-02 ERα Signaling Is Required for TrkB-Mediated Hippocampal Neuroprotection in Female Neonatal Mice after Hypoxic Ischemic Encephalopathy123 Cikla, Ulas Chanana, Vishal Kintner, Douglas B. Udho, Eshwar Eickhoff, Jens Sun, Wendy Marquez, Stephanie Covert, Lucia Otles, Arel Shapiro, Robert A. Ferrazzano, Peter Vemuganti, Raghu Levine, Jon E. Cengiz, Pelin eNeuro Theory/New Concepts Male neonate brains are more susceptible to the effects of perinatal asphyxia resulting in hypoxia and ischemia (HI)-related brain injury. The relative resistance of female neonatal brains to adverse consequences of HI suggests that there are sex-specific mechanisms that afford females greater neuroprotection and/or facilitates recovery post-HI. We hypothesized that HI preferentially induces estrogen receptor α (ERα) expression in female neonatal hippocampi and that ERα is coupled to Src family kinase (SFK) activation that in turn augments phosphorylation of the TrkB and thereby results in decreased apoptosis. After inducing the Vannucci’s HI model on P9 (C57BL/6J) mice, female and male ERα wild-type (ERα(+/+)) or ERα null mutant (ERα(−/−)) mice received vehicle control or the selective TrkB agonist 7,8-dihydroxyflavone (7,8-DHF). Hippocampi were collected for analysis of mRNA of ERα and BDNF, protein levels of ERα, p-TrkB, p-src, and cleaved caspase 3 (c-caspase-3) post-HI. Our results demonstrate that: (1) HI differentially induces ERα expression in the hippocampus of the female versus male neonate, (2) src and TrkB phosphorylation post-HI is greater in females than in males after 7,8-DHF therapy, (3) src and TrkB phosphorylation post-HI depend on the presence of ERα, and (4) TrkB agonist therapy decreases the c-caspase-3 only in ERα(+/+) female mice hippocampus. Together, these observations provide evidence that female-specific induction of ERα expression confers neuroprotection with TrkB agonist therapy via SFK activation and account for improved functional outcomes in female neonates post-HI. Society for Neuroscience 2016-01-28 /pmc/articles/PMC4731462/ /pubmed/26839918 http://dx.doi.org/10.1523/ENEURO.0025-15.2015 Text en Copyright © 2016 Cikla 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 (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 Theory/New Concepts
Cikla, Ulas
Chanana, Vishal
Kintner, Douglas B.
Udho, Eshwar
Eickhoff, Jens
Sun, Wendy
Marquez, Stephanie
Covert, Lucia
Otles, Arel
Shapiro, Robert A.
Ferrazzano, Peter
Vemuganti, Raghu
Levine, Jon E.
Cengiz, Pelin
ERα Signaling Is Required for TrkB-Mediated Hippocampal Neuroprotection in Female Neonatal Mice after Hypoxic Ischemic Encephalopathy123
title ERα Signaling Is Required for TrkB-Mediated Hippocampal Neuroprotection in Female Neonatal Mice after Hypoxic Ischemic Encephalopathy123
title_full ERα Signaling Is Required for TrkB-Mediated Hippocampal Neuroprotection in Female Neonatal Mice after Hypoxic Ischemic Encephalopathy123
title_fullStr ERα Signaling Is Required for TrkB-Mediated Hippocampal Neuroprotection in Female Neonatal Mice after Hypoxic Ischemic Encephalopathy123
title_full_unstemmed ERα Signaling Is Required for TrkB-Mediated Hippocampal Neuroprotection in Female Neonatal Mice after Hypoxic Ischemic Encephalopathy123
title_short ERα Signaling Is Required for TrkB-Mediated Hippocampal Neuroprotection in Female Neonatal Mice after Hypoxic Ischemic Encephalopathy123
title_sort erα signaling is required for trkb-mediated hippocampal neuroprotection in female neonatal mice after hypoxic ischemic encephalopathy123
topic Theory/New Concepts
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4731462/
https://www.ncbi.nlm.nih.gov/pubmed/26839918
http://dx.doi.org/10.1523/ENEURO.0025-15.2015
work_keys_str_mv AT ciklaulas erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT chananavishal erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT kintnerdouglasb erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT udhoeshwar erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT eickhoffjens erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT sunwendy erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT marquezstephanie erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT covertlucia erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT otlesarel erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT shapiroroberta erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT ferrazzanopeter erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT vemugantiraghu erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT levinejone erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123
AT cengizpelin erasignalingisrequiredfortrkbmediatedhippocampalneuroprotectioninfemaleneonatalmiceafterhypoxicischemicencephalopathy123