Tauroursodeoxycholic Acid Drives Mitochondrial Bioenergetics Toward Neural Stem Cell Proliferation

Detalhes bibliográficos
Autor(a) principal: Fernandes, Marta Sofia Barbosa
Data de Publicação: 2017
Tipo de documento: Dissertação
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: http://hdl.handle.net/10362/23872
Resumo: Neurogenesis occurs throughout life in discrete areas of the adult mammalian brain. Unfortunately, there is a lack of effective regeneration during aging or after injury. Therefore, life-long potentiation of endogenous neurogenesis represents a major issue. Curiously, proliferation and differentiation potential of neural stem cells (NSCs) were recently shown to be highly dependent on mitochondrial bioenergetics and fatty acid (FA) lipogenesis. Furthermore, tauroursodeoxycholic acid (TUDCA), an endogenous neuroprotective bile acid, considered a regulator of energy metabolism and an inhibitor of early differentiation-associated apoptosis events in NSCs, stimulates proliferation and neuronal conversion of these cells. We aimed to clarify the impact of TUDCA on the mitochondrial proteome in self-renewing or differentiating mouse NSCs, using liquid chromatography coupled with mass spectrometry (LC-MS) based detection of differential proteomics. Validation of mitochondrial proteomic analysis by Western blot in two different NSC lines revealed that TUDCA significantly decreases the mitochondrial levels of long-chain acyl-CoA dehydrogenase (LCAD) protein upon differentiation, an enzyme crucial for β-oxidation of long-chain FAs. Further, nuclear levels of sterol regulatory element-binding protein (SREBP-1), a major transcription factor of lipid biosynthesis, were also found significantly increased, as the levels of palmitic and stearic FAs raise up. Interestingly, mitochondrial levels of pyruvate dehydrogenase E1-α (PDHE1-α), an enzymatic subunit belonging to glucose metabolism, were also markedly enhanced by TUDCA. Of note, TUDCA promoted mitochondria-nucleus translocation of PDHE1-α. Therefore, the proliferative role of this bile acid may rely, in part, in increasing the pool of mitochondrial and/or nuclear acetyl-CoA to assure NSC cycle progression. Finally, LCAD, SREBP-1, and PDHE1-α expression profiles were also assessed during early stages of neural differentiation bringing novel insights to NSC metabolic choices throughout differentiation. Altogether, our results unravel the metabolic impact of TUDCA in controlling NSC fate, demonstrating that this bile acid not only induces mitochondrial advantageous conditions but also metabolic plasticity.
id RCAP_9c87f6687b3cfb3ca5ee1849be63f4a3
oai_identifier_str oai:run.unl.pt:10362/23872
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str 7160
spelling Tauroursodeoxycholic Acid Drives Mitochondrial Bioenergetics Toward Neural Stem Cell ProliferationDifferentiationLipid metabolismMitochondriaNeural stem cellsProliferationTauroursodeoxycholic acidDomínio/Área Científica::Engenharia e Tecnologia::Outras Engenharias e TecnologiasNeurogenesis occurs throughout life in discrete areas of the adult mammalian brain. Unfortunately, there is a lack of effective regeneration during aging or after injury. Therefore, life-long potentiation of endogenous neurogenesis represents a major issue. Curiously, proliferation and differentiation potential of neural stem cells (NSCs) were recently shown to be highly dependent on mitochondrial bioenergetics and fatty acid (FA) lipogenesis. Furthermore, tauroursodeoxycholic acid (TUDCA), an endogenous neuroprotective bile acid, considered a regulator of energy metabolism and an inhibitor of early differentiation-associated apoptosis events in NSCs, stimulates proliferation and neuronal conversion of these cells. We aimed to clarify the impact of TUDCA on the mitochondrial proteome in self-renewing or differentiating mouse NSCs, using liquid chromatography coupled with mass spectrometry (LC-MS) based detection of differential proteomics. Validation of mitochondrial proteomic analysis by Western blot in two different NSC lines revealed that TUDCA significantly decreases the mitochondrial levels of long-chain acyl-CoA dehydrogenase (LCAD) protein upon differentiation, an enzyme crucial for β-oxidation of long-chain FAs. Further, nuclear levels of sterol regulatory element-binding protein (SREBP-1), a major transcription factor of lipid biosynthesis, were also found significantly increased, as the levels of palmitic and stearic FAs raise up. Interestingly, mitochondrial levels of pyruvate dehydrogenase E1-α (PDHE1-α), an enzymatic subunit belonging to glucose metabolism, were also markedly enhanced by TUDCA. Of note, TUDCA promoted mitochondria-nucleus translocation of PDHE1-α. Therefore, the proliferative role of this bile acid may rely, in part, in increasing the pool of mitochondrial and/or nuclear acetyl-CoA to assure NSC cycle progression. Finally, LCAD, SREBP-1, and PDHE1-α expression profiles were also assessed during early stages of neural differentiation bringing novel insights to NSC metabolic choices throughout differentiation. Altogether, our results unravel the metabolic impact of TUDCA in controlling NSC fate, demonstrating that this bile acid not only induces mitochondrial advantageous conditions but also metabolic plasticity.Solá, SusanaRodrigues, CecíliaRUNFernandes, Marta Sofia Barbosa2019-10-01T00:30:32Z2017-092017-092017-09-01T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/masterThesisapplication/pdfhttp://hdl.handle.net/10362/23872enginfo:eu-repo/semantics/embargoedAccessreponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãoinstacron:RCAAP2024-03-11T04:12:17Zoai:run.unl.pt:10362/23872Portal AgregadorONGhttps://www.rcaap.pt/oai/openaireopendoar:71602024-03-20T03:27:55.855064Repositório 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 Tauroursodeoxycholic Acid Drives Mitochondrial Bioenergetics Toward Neural Stem Cell Proliferation
title Tauroursodeoxycholic Acid Drives Mitochondrial Bioenergetics Toward Neural Stem Cell Proliferation
spellingShingle Tauroursodeoxycholic Acid Drives Mitochondrial Bioenergetics Toward Neural Stem Cell Proliferation
Fernandes, Marta Sofia Barbosa
Differentiation
Lipid metabolism
Mitochondria
Neural stem cells
Proliferation
Tauroursodeoxycholic acid
Domínio/Área Científica::Engenharia e Tecnologia::Outras Engenharias e Tecnologias
title_short Tauroursodeoxycholic Acid Drives Mitochondrial Bioenergetics Toward Neural Stem Cell Proliferation
title_full Tauroursodeoxycholic Acid Drives Mitochondrial Bioenergetics Toward Neural Stem Cell Proliferation
title_fullStr Tauroursodeoxycholic Acid Drives Mitochondrial Bioenergetics Toward Neural Stem Cell Proliferation
title_full_unstemmed Tauroursodeoxycholic Acid Drives Mitochondrial Bioenergetics Toward Neural Stem Cell Proliferation
title_sort Tauroursodeoxycholic Acid Drives Mitochondrial Bioenergetics Toward Neural Stem Cell Proliferation
author Fernandes, Marta Sofia Barbosa
author_facet Fernandes, Marta Sofia Barbosa
author_role author
dc.contributor.none.fl_str_mv Solá, Susana
Rodrigues, Cecília
RUN
dc.contributor.author.fl_str_mv Fernandes, Marta Sofia Barbosa
dc.subject.por.fl_str_mv Differentiation
Lipid metabolism
Mitochondria
Neural stem cells
Proliferation
Tauroursodeoxycholic acid
Domínio/Área Científica::Engenharia e Tecnologia::Outras Engenharias e Tecnologias
topic Differentiation
Lipid metabolism
Mitochondria
Neural stem cells
Proliferation
Tauroursodeoxycholic acid
Domínio/Área Científica::Engenharia e Tecnologia::Outras Engenharias e Tecnologias
description Neurogenesis occurs throughout life in discrete areas of the adult mammalian brain. Unfortunately, there is a lack of effective regeneration during aging or after injury. Therefore, life-long potentiation of endogenous neurogenesis represents a major issue. Curiously, proliferation and differentiation potential of neural stem cells (NSCs) were recently shown to be highly dependent on mitochondrial bioenergetics and fatty acid (FA) lipogenesis. Furthermore, tauroursodeoxycholic acid (TUDCA), an endogenous neuroprotective bile acid, considered a regulator of energy metabolism and an inhibitor of early differentiation-associated apoptosis events in NSCs, stimulates proliferation and neuronal conversion of these cells. We aimed to clarify the impact of TUDCA on the mitochondrial proteome in self-renewing or differentiating mouse NSCs, using liquid chromatography coupled with mass spectrometry (LC-MS) based detection of differential proteomics. Validation of mitochondrial proteomic analysis by Western blot in two different NSC lines revealed that TUDCA significantly decreases the mitochondrial levels of long-chain acyl-CoA dehydrogenase (LCAD) protein upon differentiation, an enzyme crucial for β-oxidation of long-chain FAs. Further, nuclear levels of sterol regulatory element-binding protein (SREBP-1), a major transcription factor of lipid biosynthesis, were also found significantly increased, as the levels of palmitic and stearic FAs raise up. Interestingly, mitochondrial levels of pyruvate dehydrogenase E1-α (PDHE1-α), an enzymatic subunit belonging to glucose metabolism, were also markedly enhanced by TUDCA. Of note, TUDCA promoted mitochondria-nucleus translocation of PDHE1-α. Therefore, the proliferative role of this bile acid may rely, in part, in increasing the pool of mitochondrial and/or nuclear acetyl-CoA to assure NSC cycle progression. Finally, LCAD, SREBP-1, and PDHE1-α expression profiles were also assessed during early stages of neural differentiation bringing novel insights to NSC metabolic choices throughout differentiation. Altogether, our results unravel the metabolic impact of TUDCA in controlling NSC fate, demonstrating that this bile acid not only induces mitochondrial advantageous conditions but also metabolic plasticity.
publishDate 2017
dc.date.none.fl_str_mv 2017-09
2017-09
2017-09-01T00:00:00Z
2019-10-01T00:30:32Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/masterThesis
format masterThesis
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://hdl.handle.net/10362/23872
url http://hdl.handle.net/10362/23872
dc.language.iso.fl_str_mv eng
language eng
dc.rights.driver.fl_str_mv info:eu-repo/semantics/embargoedAccess
eu_rights_str_mv embargoedAccess
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_ 1799137905951637504