HAT cofactor TRRAP modulates microtubule dynamics via SP1 signaling to prevent neurodegeneration

Brain homeostasis is regulated by the viability and functionality of neurons. HAT (histone acetyltransferase) and HDAC (histone deacetylase) inhibitors have been applied to treat neurological deficits in humans; yet, the epigenetic regulation in neurodegeneration remains elusive. Mutations of HAT co...

Full description

Bibliographic Details
Main Authors: Alicia Tapias, David Lázaro, Bo-Kun Yin, Seyed Mohammad Mahdi Rasa, Anna Krepelova, Erika Kelmer Sacramento, Paulius Grigaravicius, Philipp Koch, Joanna Kirkpatrick, Alessandro Ori, Francesco Neri, Zhao-Qi Wang
Format: Article
Language:English
Published: eLife Sciences Publications Ltd 2021-02-01
Series:eLife
Subjects:
SP1
Online Access:https://elifesciences.org/articles/61531
id doaj-33e304c7257749b2a06dea0c9d2f2260
record_format Article
spelling doaj-33e304c7257749b2a06dea0c9d2f22602021-07-19T11:51:45ZengeLife Sciences Publications LtdeLife2050-084X2021-02-011010.7554/eLife.61531HAT cofactor TRRAP modulates microtubule dynamics via SP1 signaling to prevent neurodegenerationAlicia Tapias0David Lázaro1Bo-Kun Yin2Seyed Mohammad Mahdi Rasa3https://orcid.org/0000-0001-6850-8909Anna Krepelova4Erika Kelmer Sacramento5Paulius Grigaravicius6Philipp Koch7https://orcid.org/0000-0003-2825-7943Joanna Kirkpatrick8Alessandro Ori9https://orcid.org/0000-0002-3046-0871Francesco Neri10Zhao-Qi Wang11https://orcid.org/0000-0002-8336-3485Leibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, GermanyLeibniz Institute on Aging – Fritz Lipmann Institute (FLI), Jena, Germany; Faculty of Biological Sciences, Friedrich-Schiller-University of Jena, Jena, GermanyBrain homeostasis is regulated by the viability and functionality of neurons. HAT (histone acetyltransferase) and HDAC (histone deacetylase) inhibitors have been applied to treat neurological deficits in humans; yet, the epigenetic regulation in neurodegeneration remains elusive. Mutations of HAT cofactor TRRAP (transformation/transcription domain-associated protein) cause human neuropathies, including psychosis, intellectual disability, autism, and epilepsy, with unknown mechanism. Here we show that Trrap deletion in Purkinje neurons results in neurodegeneration of old mice. Integrated transcriptomics, epigenomics, and proteomics reveal that TRRAP via SP1 conducts a conserved transcriptomic program. TRRAP is required for SP1 binding at the promoter proximity of target genes, especially microtubule dynamics. The ectopic expression of Stathmin3/4 ameliorates defects of TRRAP-deficient neurons, indicating that the microtubule dynamics is particularly vulnerable to the action of SP1 activity. This study unravels a network linking three well-known, but up-to-date unconnected, signaling pathways, namely TRRAP, HAT, and SP1 with microtubule dynamics, in neuroprotection.https://elifesciences.org/articles/61531cell linesTRRAPSP1Brainstathmins
collection DOAJ
language English
format Article
sources DOAJ
author Alicia Tapias
David Lázaro
Bo-Kun Yin
Seyed Mohammad Mahdi Rasa
Anna Krepelova
Erika Kelmer Sacramento
Paulius Grigaravicius
Philipp Koch
Joanna Kirkpatrick
Alessandro Ori
Francesco Neri
Zhao-Qi Wang
spellingShingle Alicia Tapias
David Lázaro
Bo-Kun Yin
Seyed Mohammad Mahdi Rasa
Anna Krepelova
Erika Kelmer Sacramento
Paulius Grigaravicius
Philipp Koch
Joanna Kirkpatrick
Alessandro Ori
Francesco Neri
Zhao-Qi Wang
HAT cofactor TRRAP modulates microtubule dynamics via SP1 signaling to prevent neurodegeneration
eLife
cell lines
TRRAP
SP1
Brain
stathmins
author_facet Alicia Tapias
David Lázaro
Bo-Kun Yin
Seyed Mohammad Mahdi Rasa
Anna Krepelova
Erika Kelmer Sacramento
Paulius Grigaravicius
Philipp Koch
Joanna Kirkpatrick
Alessandro Ori
Francesco Neri
Zhao-Qi Wang
author_sort Alicia Tapias
title HAT cofactor TRRAP modulates microtubule dynamics via SP1 signaling to prevent neurodegeneration
title_short HAT cofactor TRRAP modulates microtubule dynamics via SP1 signaling to prevent neurodegeneration
title_full HAT cofactor TRRAP modulates microtubule dynamics via SP1 signaling to prevent neurodegeneration
title_fullStr HAT cofactor TRRAP modulates microtubule dynamics via SP1 signaling to prevent neurodegeneration
title_full_unstemmed HAT cofactor TRRAP modulates microtubule dynamics via SP1 signaling to prevent neurodegeneration
title_sort hat cofactor trrap modulates microtubule dynamics via sp1 signaling to prevent neurodegeneration
publisher eLife Sciences Publications Ltd
series eLife
issn 2050-084X
publishDate 2021-02-01
description Brain homeostasis is regulated by the viability and functionality of neurons. HAT (histone acetyltransferase) and HDAC (histone deacetylase) inhibitors have been applied to treat neurological deficits in humans; yet, the epigenetic regulation in neurodegeneration remains elusive. Mutations of HAT cofactor TRRAP (transformation/transcription domain-associated protein) cause human neuropathies, including psychosis, intellectual disability, autism, and epilepsy, with unknown mechanism. Here we show that Trrap deletion in Purkinje neurons results in neurodegeneration of old mice. Integrated transcriptomics, epigenomics, and proteomics reveal that TRRAP via SP1 conducts a conserved transcriptomic program. TRRAP is required for SP1 binding at the promoter proximity of target genes, especially microtubule dynamics. The ectopic expression of Stathmin3/4 ameliorates defects of TRRAP-deficient neurons, indicating that the microtubule dynamics is particularly vulnerable to the action of SP1 activity. This study unravels a network linking three well-known, but up-to-date unconnected, signaling pathways, namely TRRAP, HAT, and SP1 with microtubule dynamics, in neuroprotection.
topic cell lines
TRRAP
SP1
Brain
stathmins
url https://elifesciences.org/articles/61531
work_keys_str_mv AT aliciatapias hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT davidlazaro hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT bokunyin hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT seyedmohammadmahdirasa hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT annakrepelova hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT erikakelmersacramento hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT pauliusgrigaravicius hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT philippkoch hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT joannakirkpatrick hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT alessandroori hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT francesconeri hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
AT zhaoqiwang hatcofactortrrapmodulatesmicrotubuledynamicsviasp1signalingtopreventneurodegeneration
_version_ 1721294963040321536