Cargando…

Seizure‐induced impairment in neuronal ketogenesis: Role of zinc‐α2‐glycoprotein in mitochondria

Ketone bodies (KBs) were known to suppress seizure. Untraditionally, neurons were recently reported to utilize fatty acids and produce KBs, but the effect of seizure on neuronal ketogenesis has not been researched. Zinc‐α2‐glycoprotein (ZAG) was reported to suppress seizure via unclear mechanism. In...

Descripción completa

Detalles Bibliográficos
Autores principales: Tan, Changhong, Liu, Xi, Peng, Wuxue, Wang, Hui, Zhou, Wen, Jiang, Jin, Wei, Xin, Mo, Lijuan, Chen, Yangmei, Chen, Lifen
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7299723/
https://www.ncbi.nlm.nih.gov/pubmed/32340079
http://dx.doi.org/10.1111/jcmm.15337
_version_ 1783547448080203776
author Tan, Changhong
Liu, Xi
Peng, Wuxue
Wang, Hui
Zhou, Wen
Jiang, Jin
Wei, Xin
Mo, Lijuan
Chen, Yangmei
Chen, Lifen
author_facet Tan, Changhong
Liu, Xi
Peng, Wuxue
Wang, Hui
Zhou, Wen
Jiang, Jin
Wei, Xin
Mo, Lijuan
Chen, Yangmei
Chen, Lifen
author_sort Tan, Changhong
collection PubMed
description Ketone bodies (KBs) were known to suppress seizure. Untraditionally, neurons were recently reported to utilize fatty acids and produce KBs, but the effect of seizure on neuronal ketogenesis has not been researched. Zinc‐α2‐glycoprotein (ZAG) was reported to suppress seizure via unclear mechanism. Interestingly, ZAG was involved in fatty acid β‐oxidation and thus may exert anti‐epileptic effect by promoting ketogenesis. However, this promotive effect of ZAG on neuronal ketogenesis has not been clarified. In this study, we performed immunoprecipitation and mass spectrometry to identify potential interaction partners with ZAG. The mechanisms of how ZAG translocated into mitochondria were determined by quantitative coimmunoprecipitation after treatment with apoptozole, a heat shock cognate protein 70 (HSC70) inhibitor. ZAG level was modulated by lentivirus in neurons or adeno‐associated virus in rat brains. Seizure models were induced by magnesium (Mg(2+))‐free artificial cerebrospinal fluid in neurons or intraperitoneal injection of pentylenetetrazole kindling in rats. Ketogenesis was determined by cyclic thio‐NADH method in supernatant of neurons or brain homogenate. The effect of peroxisome proliferator–activated receptor γ (PPARγ) on ZAG expression was examined by Western blot, quantitative real‐time polymerase chain reaction (qRT‐PCR) and chromatin immunoprecipitation qRT‐PCR. We found that seizure induced ketogenesis deficiency via a ZAG‐dependent mechanism. ZAG entered mitochondria through a HSC70‐dependent mechanism, promoted ketogenesis by binding to four β‐subunits of long‐chain L‐3‐hydroxyacyl‐CoA dehydrogenase (HADHB) and alleviated ketogenesis impairment in a neuronal seizure model and pentylenetetrazole‐kindled epileptic rats. Additionally, PPARγ activation up‐regulated ZAG expression by binding to promoter region of AZGP1 gene and promoted ketogenesis through a ZAG‐dependent mechanism.
format Online
Article
Text
id pubmed-7299723
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-72997232020-06-18 Seizure‐induced impairment in neuronal ketogenesis: Role of zinc‐α2‐glycoprotein in mitochondria Tan, Changhong Liu, Xi Peng, Wuxue Wang, Hui Zhou, Wen Jiang, Jin Wei, Xin Mo, Lijuan Chen, Yangmei Chen, Lifen J Cell Mol Med Original Articles Ketone bodies (KBs) were known to suppress seizure. Untraditionally, neurons were recently reported to utilize fatty acids and produce KBs, but the effect of seizure on neuronal ketogenesis has not been researched. Zinc‐α2‐glycoprotein (ZAG) was reported to suppress seizure via unclear mechanism. Interestingly, ZAG was involved in fatty acid β‐oxidation and thus may exert anti‐epileptic effect by promoting ketogenesis. However, this promotive effect of ZAG on neuronal ketogenesis has not been clarified. In this study, we performed immunoprecipitation and mass spectrometry to identify potential interaction partners with ZAG. The mechanisms of how ZAG translocated into mitochondria were determined by quantitative coimmunoprecipitation after treatment with apoptozole, a heat shock cognate protein 70 (HSC70) inhibitor. ZAG level was modulated by lentivirus in neurons or adeno‐associated virus in rat brains. Seizure models were induced by magnesium (Mg(2+))‐free artificial cerebrospinal fluid in neurons or intraperitoneal injection of pentylenetetrazole kindling in rats. Ketogenesis was determined by cyclic thio‐NADH method in supernatant of neurons or brain homogenate. The effect of peroxisome proliferator–activated receptor γ (PPARγ) on ZAG expression was examined by Western blot, quantitative real‐time polymerase chain reaction (qRT‐PCR) and chromatin immunoprecipitation qRT‐PCR. We found that seizure induced ketogenesis deficiency via a ZAG‐dependent mechanism. ZAG entered mitochondria through a HSC70‐dependent mechanism, promoted ketogenesis by binding to four β‐subunits of long‐chain L‐3‐hydroxyacyl‐CoA dehydrogenase (HADHB) and alleviated ketogenesis impairment in a neuronal seizure model and pentylenetetrazole‐kindled epileptic rats. Additionally, PPARγ activation up‐regulated ZAG expression by binding to promoter region of AZGP1 gene and promoted ketogenesis through a ZAG‐dependent mechanism. John Wiley and Sons Inc. 2020-04-27 2020-06 /pmc/articles/PMC7299723/ /pubmed/32340079 http://dx.doi.org/10.1111/jcmm.15337 Text en © 2020 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine. This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Articles
Tan, Changhong
Liu, Xi
Peng, Wuxue
Wang, Hui
Zhou, Wen
Jiang, Jin
Wei, Xin
Mo, Lijuan
Chen, Yangmei
Chen, Lifen
Seizure‐induced impairment in neuronal ketogenesis: Role of zinc‐α2‐glycoprotein in mitochondria
title Seizure‐induced impairment in neuronal ketogenesis: Role of zinc‐α2‐glycoprotein in mitochondria
title_full Seizure‐induced impairment in neuronal ketogenesis: Role of zinc‐α2‐glycoprotein in mitochondria
title_fullStr Seizure‐induced impairment in neuronal ketogenesis: Role of zinc‐α2‐glycoprotein in mitochondria
title_full_unstemmed Seizure‐induced impairment in neuronal ketogenesis: Role of zinc‐α2‐glycoprotein in mitochondria
title_short Seizure‐induced impairment in neuronal ketogenesis: Role of zinc‐α2‐glycoprotein in mitochondria
title_sort seizure‐induced impairment in neuronal ketogenesis: role of zinc‐α2‐glycoprotein in mitochondria
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7299723/
https://www.ncbi.nlm.nih.gov/pubmed/32340079
http://dx.doi.org/10.1111/jcmm.15337
work_keys_str_mv AT tanchanghong seizureinducedimpairmentinneuronalketogenesisroleofzinca2glycoproteininmitochondria
AT liuxi seizureinducedimpairmentinneuronalketogenesisroleofzinca2glycoproteininmitochondria
AT pengwuxue seizureinducedimpairmentinneuronalketogenesisroleofzinca2glycoproteininmitochondria
AT wanghui seizureinducedimpairmentinneuronalketogenesisroleofzinca2glycoproteininmitochondria
AT zhouwen seizureinducedimpairmentinneuronalketogenesisroleofzinca2glycoproteininmitochondria
AT jiangjin seizureinducedimpairmentinneuronalketogenesisroleofzinca2glycoproteininmitochondria
AT weixin seizureinducedimpairmentinneuronalketogenesisroleofzinca2glycoproteininmitochondria
AT molijuan seizureinducedimpairmentinneuronalketogenesisroleofzinca2glycoproteininmitochondria
AT chenyangmei seizureinducedimpairmentinneuronalketogenesisroleofzinca2glycoproteininmitochondria
AT chenlifen seizureinducedimpairmentinneuronalketogenesisroleofzinca2glycoproteininmitochondria