Show simple item record

Files in this item

Thumbnail

Item metadata

dc.contributor.authorMetodieva, Vanya
dc.contributor.authorSmith, Terry Kenneth
dc.contributor.authorGunn-Moore, Frank James
dc.date.accessioned2022-10-17T15:30:02Z
dc.date.available2022-10-17T15:30:02Z
dc.date.issued2022-09-12
dc.identifier281524112
dc.identifier6e6978d9-cff9-4de7-9a5c-c29958b50362
dc.identifier36096650
dc.identifier85139377301
dc.identifier000869099900004
dc.identifier.citationMetodieva , V , Smith , T K & Gunn-Moore , F J 2022 , ' The mitochondrial enzyme 17βHSD10 modulates ischemic and amyloid-β-induced stress in primary mouse astrocytes ' , eNeuro , vol. 9 , no. 5 . https://doi.org/10.1523/ENEURO.0040-22.2022en
dc.identifier.issn2373-2822
dc.identifier.otherJisc: 623285
dc.identifier.otherpii: ENEURO.0040-22.2022
dc.identifier.otherORCID: /0000-0003-3422-3387/work/120051526
dc.identifier.urihttps://hdl.handle.net/10023/26206
dc.descriptionFunding information: The project is supported by Alzheimer’s Society and RS Macdonald Charitable Trust.en
dc.description.abstractSevere brain metabolic dysfunction and amyloid-beta accumulation are key hallmarks of Alzheimer's disease. While astrocytes contribute to both pathological mechanisms, the role of their mitochondria, which is essential for signalling and maintenance of these processes, has been largely understudied. The current work provides the first direct evidence that the mitochondrial metabolic switch 17β-hydroxysteroid dehydrogenase type 10 (17βHSD10) is expressed and active in murine astrocytes from different brain regions. While it is known that this protein is overexpressed in the brains of Alzheimer's disease patients, we found that 17βHSD10 is also upregulated in astrocytes exposed to amyloidogenic and ischemic stress. Importantly, such catalytic overexpression of 17βHSD10 inhibits mitochondrial respiration during increased energy demand. This observation contrasts with what has been found in neuronal and cancer model systems, which suggests astrocyte-specific mechanisms mediated by the protein. Furthermore, the catalytic upregulation of the enzyme exacerbates astrocytic damage, reactive oxygen species generation and mitochondrial network alterations during amyloidogenic stress. On the other hand, 17βHSD10 inhibition through AG18051 counters most of these effects. In conclusion, our data represents novel insights into the role of astrocytic mitochondria in metabolic and amyloidogenic stress with implications of 17βHSD10 in multiple neurodegenerative mechanisms. The current study presents the first direct evidence for the role of enzymatic activity of 17βHSD10 in astrocytes. We report that the protein is involved in the response of cortical astrocytes to stress conditions associated with ischemic stroke and AD. Furthermore, 17βHSD10 regulates astrocytic mitochondrial function, and the effects differ from what has been reported in neurons. These findings contribute to a growing body of evidence showing that astrocytic mitochondria are a key factor in neurodegenerative pathology. Considering the 17βHSD10-targeting therapeutics currently being developed for AD, these findings provide important insight into the role of this target in the cell population which carries out key metabolic support and toxic clearance from the brain.
dc.format.extent24
dc.format.extent4809850
dc.language.isoeng
dc.relation.ispartofeNeuroen
dc.subjectIschemiaen
dc.subjectAlzheimer’s diseaseen
dc.subjectAstrocytesen
dc.subjectHSD10)en
dc.subjectMitochondriaen
dc.subject17β-hydroxysteroid dehydrogenase type 10 (17β-en
dc.subjectMetabolismen
dc.subjectRC0321 Neuroscience. Biological psychiatry. Neuropsychiatryen
dc.subject3rd-DASen
dc.subjectSDG 3 - Good Health and Well-beingen
dc.subjectMCCen
dc.subject.lccRC0321en
dc.titleThe mitochondrial enzyme 17βHSD10 modulates ischemic and amyloid-β-induced stress in primary mouse astrocytesen
dc.typeJournal articleen
dc.contributor.institutionUniversity of St Andrews. School of Biologyen
dc.contributor.institutionUniversity of St Andrews. Sir James Mackenzie Institute for Early Diagnosisen
dc.contributor.institutionUniversity of St Andrews. Biomedical Sciences Research Complexen
dc.contributor.institutionUniversity of St Andrews. Centre for Biophotonicsen
dc.contributor.institutionUniversity of St Andrews. Institute of Behavioural and Neural Sciencesen
dc.identifier.doi10.1523/ENEURO.0040-22.2022
dc.description.statusPeer revieweden


This item appears in the following Collection(s)

Show simple item record