Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after stroke

Detalhes bibliográficos
Autor(a) principal: Talhada, Daniela
Data de Publicação: 2019
Outros Autores: Feiteiro, Joana, Costa, Ana Raquel, Talhada, Tiago, Cairrão, Elisa, Wieloch, Tadeusz, Englund, Elisabet, Santos, Cecilia Reis, Gonçalves, Isabel, Ruscher, Karsten
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: http://hdl.handle.net/10400.6/9648
Resumo: The development of new therapeutic approaches for stroke patients requires a detailed understanding of the mechanisms that enhance recovery of lost neurological functions. The efficacy to enhance homeostatic mechanisms during the first weeks after stroke will influence functional outcome. Thyroid hormones (TH) are essential regulators of neuronal plasticity, however, their role in recovery related mechanisms of neuronal plasticity after stroke remains unknown. This study addresses important findings of 3,5,3'-triiodo-L-thyronine (T3) in the regulation of homeostatic mechanisms that adjust excitability - inhibition ratio in the post-ischemic brain. This is valid during the first 2 weeks after experimental stroke induced by photothrombosis (PT) and in cultured neurons subjected to an in vitro model of acute cerebral ischemia. In the human post-stroke brain, we assessed the expression pattern of TH receptors (TR) protein levels, important for mediating T3 actions.Our results show that T3 modulates several plasticity mechanisms that may operate on different temporal and spatial scales as compensatory mechanisms to assure appropriate synaptic neurotransmission. We have shown in vivo that long-term administration of T3 after PT significantly (1) enhances lost sensorimotor function; (2) increases levels of synaptotagmin 1&2 and levels of the post-synaptic GluR2 subunit in AMPA receptors in the peri-infarct area; (3) increases dendritic spine density in the peri-infarct and contralateral region and (4) decreases tonic GABAergic signaling in the peri-infarct area by a reduced number of parvalbumin+ / c-fos+ neurons and glutamic acid decarboxylase 65/67 levels. In addition, we have shown that T3 modulates in vitro neuron membrane properties with the balance of inward glutamate ligand-gated channels currents and decreases synaptotagmin levels in conditions of deprived oxygen and glucose. Interestingly, we found increased levels of TRβ1 in the infarct core of post-mortem human stroke patients, which mediate T3 actions. Summarizing, our data identify T3 as a potential key therapeutic agent to enhance recovery of lost neurological functions after ischemic stroke.
id RCAP_95dc2e33d7917b51faa15a616b7cb586
oai_identifier_str oai:ubibliorum.ubi.pt:10400.6/9648
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str 7160
spelling Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after strokeIschemiaPhotothrombosisRecoveryStrokeThyroid hormonesThyroid hormone receptorsThe development of new therapeutic approaches for stroke patients requires a detailed understanding of the mechanisms that enhance recovery of lost neurological functions. The efficacy to enhance homeostatic mechanisms during the first weeks after stroke will influence functional outcome. Thyroid hormones (TH) are essential regulators of neuronal plasticity, however, their role in recovery related mechanisms of neuronal plasticity after stroke remains unknown. This study addresses important findings of 3,5,3'-triiodo-L-thyronine (T3) in the regulation of homeostatic mechanisms that adjust excitability - inhibition ratio in the post-ischemic brain. This is valid during the first 2 weeks after experimental stroke induced by photothrombosis (PT) and in cultured neurons subjected to an in vitro model of acute cerebral ischemia. In the human post-stroke brain, we assessed the expression pattern of TH receptors (TR) protein levels, important for mediating T3 actions.Our results show that T3 modulates several plasticity mechanisms that may operate on different temporal and spatial scales as compensatory mechanisms to assure appropriate synaptic neurotransmission. We have shown in vivo that long-term administration of T3 after PT significantly (1) enhances lost sensorimotor function; (2) increases levels of synaptotagmin 1&2 and levels of the post-synaptic GluR2 subunit in AMPA receptors in the peri-infarct area; (3) increases dendritic spine density in the peri-infarct and contralateral region and (4) decreases tonic GABAergic signaling in the peri-infarct area by a reduced number of parvalbumin+ / c-fos+ neurons and glutamic acid decarboxylase 65/67 levels. In addition, we have shown that T3 modulates in vitro neuron membrane properties with the balance of inward glutamate ligand-gated channels currents and decreases synaptotagmin levels in conditions of deprived oxygen and glucose. Interestingly, we found increased levels of TRβ1 in the infarct core of post-mortem human stroke patients, which mediate T3 actions. Summarizing, our data identify T3 as a potential key therapeutic agent to enhance recovery of lost neurological functions after ischemic stroke.uBibliorumTalhada, DanielaFeiteiro, JoanaCosta, Ana RaquelTalhada, TiagoCairrão, ElisaWieloch, TadeuszEnglund, ElisabetSantos, Cecilia ReisGonçalves, IsabelRuscher, Karsten2020-03-02T10:51:46Z2019-12-212019-12-21T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articleapplication/pdfhttp://hdl.handle.net/10400.6/9648eng10.1186/s40478-019-0866-4info:eu-repo/semantics/openAccessreponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãoinstacron:RCAAP2023-12-15T09:49:58Zoai:ubibliorum.ubi.pt:10400.6/9648Portal AgregadorONGhttps://www.rcaap.pt/oai/openaireopendoar:71602024-03-20T00:49:22.999380Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãofalse
dc.title.none.fl_str_mv Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after stroke
title Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after stroke
spellingShingle Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after stroke
Talhada, Daniela
Ischemia
Photothrombosis
Recovery
Stroke
Thyroid hormones
Thyroid hormone receptors
title_short Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after stroke
title_full Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after stroke
title_fullStr Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after stroke
title_full_unstemmed Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after stroke
title_sort Triiodothyronine modulates neuronal plasticity mechanisms to enhance functional outcome after stroke
author Talhada, Daniela
author_facet Talhada, Daniela
Feiteiro, Joana
Costa, Ana Raquel
Talhada, Tiago
Cairrão, Elisa
Wieloch, Tadeusz
Englund, Elisabet
Santos, Cecilia Reis
Gonçalves, Isabel
Ruscher, Karsten
author_role author
author2 Feiteiro, Joana
Costa, Ana Raquel
Talhada, Tiago
Cairrão, Elisa
Wieloch, Tadeusz
Englund, Elisabet
Santos, Cecilia Reis
Gonçalves, Isabel
Ruscher, Karsten
author2_role author
author
author
author
author
author
author
author
author
dc.contributor.none.fl_str_mv uBibliorum
dc.contributor.author.fl_str_mv Talhada, Daniela
Feiteiro, Joana
Costa, Ana Raquel
Talhada, Tiago
Cairrão, Elisa
Wieloch, Tadeusz
Englund, Elisabet
Santos, Cecilia Reis
Gonçalves, Isabel
Ruscher, Karsten
dc.subject.por.fl_str_mv Ischemia
Photothrombosis
Recovery
Stroke
Thyroid hormones
Thyroid hormone receptors
topic Ischemia
Photothrombosis
Recovery
Stroke
Thyroid hormones
Thyroid hormone receptors
description The development of new therapeutic approaches for stroke patients requires a detailed understanding of the mechanisms that enhance recovery of lost neurological functions. The efficacy to enhance homeostatic mechanisms during the first weeks after stroke will influence functional outcome. Thyroid hormones (TH) are essential regulators of neuronal plasticity, however, their role in recovery related mechanisms of neuronal plasticity after stroke remains unknown. This study addresses important findings of 3,5,3'-triiodo-L-thyronine (T3) in the regulation of homeostatic mechanisms that adjust excitability - inhibition ratio in the post-ischemic brain. This is valid during the first 2 weeks after experimental stroke induced by photothrombosis (PT) and in cultured neurons subjected to an in vitro model of acute cerebral ischemia. In the human post-stroke brain, we assessed the expression pattern of TH receptors (TR) protein levels, important for mediating T3 actions.Our results show that T3 modulates several plasticity mechanisms that may operate on different temporal and spatial scales as compensatory mechanisms to assure appropriate synaptic neurotransmission. We have shown in vivo that long-term administration of T3 after PT significantly (1) enhances lost sensorimotor function; (2) increases levels of synaptotagmin 1&2 and levels of the post-synaptic GluR2 subunit in AMPA receptors in the peri-infarct area; (3) increases dendritic spine density in the peri-infarct and contralateral region and (4) decreases tonic GABAergic signaling in the peri-infarct area by a reduced number of parvalbumin+ / c-fos+ neurons and glutamic acid decarboxylase 65/67 levels. In addition, we have shown that T3 modulates in vitro neuron membrane properties with the balance of inward glutamate ligand-gated channels currents and decreases synaptotagmin levels in conditions of deprived oxygen and glucose. Interestingly, we found increased levels of TRβ1 in the infarct core of post-mortem human stroke patients, which mediate T3 actions. Summarizing, our data identify T3 as a potential key therapeutic agent to enhance recovery of lost neurological functions after ischemic stroke.
publishDate 2019
dc.date.none.fl_str_mv 2019-12-21
2019-12-21T00:00:00Z
2020-03-02T10:51:46Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/article
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://hdl.handle.net/10400.6/9648
url http://hdl.handle.net/10400.6/9648
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv 10.1186/s40478-019-0866-4
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.source.none.fl_str_mv reponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron:RCAAP
instname_str Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron_str RCAAP
institution RCAAP
reponame_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
collection Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository.name.fl_str_mv Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
repository.mail.fl_str_mv
_version_ 1799136386365784064