Interleukin-1β drives NEDD8 nuclear-to-cytoplasmic translocation, fostering parkin activation via NEDD8 binding to the P-ubiquitin activating site

Abstract Background Neuroinflammation, typified by elevated levels of interleukin-1 (IL-1) α and β, and deficits in proteostasis, characterized by accumulation of polyubiquitinated proteins and other aggregates, are associated with neurodegenerative disease independently and through interactions of...

Full description

Bibliographic Details
Main Authors: Meenakshisundaram Balasubramaniam, Paul A. Parcon, Chhanda Bose, Ling Liu, Richard A. Jones, Martin R. Farlow, Robert E. Mrak, Steven W. Barger, W. Sue T. Griffin
Format: Article
Language:English
Published: BMC 2019-12-01
Series:Journal of Neuroinflammation
Subjects:
Online Access:https://doi.org/10.1186/s12974-019-1669-z
id doaj-1228e585af3a4aaaad3af58fadaef37e
record_format Article
spelling doaj-1228e585af3a4aaaad3af58fadaef37e2020-12-27T12:06:01ZengBMCJournal of Neuroinflammation1742-20942019-12-0116111710.1186/s12974-019-1669-zInterleukin-1β drives NEDD8 nuclear-to-cytoplasmic translocation, fostering parkin activation via NEDD8 binding to the P-ubiquitin activating siteMeenakshisundaram Balasubramaniam0Paul A. Parcon1Chhanda Bose2Ling Liu3Richard A. Jones4Martin R. Farlow5Robert E. Mrak6Steven W. Barger7W. Sue T. Griffin8Department of Geriatrics, University of Arkansas for Medical SciencesDepartment of Geriatrics, University of Arkansas for Medical SciencesDepartment of Geriatrics, University of Arkansas for Medical SciencesDepartment of Geriatrics, University of Arkansas for Medical SciencesDepartment of Geriatrics, University of Arkansas for Medical SciencesDepartment of Neurology, Indiana Alzheimer Disease Center, Indiana UniversityDepartment of Pathology, University of Toledo Health Sciences CampusDepartment of Geriatrics, University of Arkansas for Medical SciencesDepartment of Geriatrics, University of Arkansas for Medical SciencesAbstract Background Neuroinflammation, typified by elevated levels of interleukin-1 (IL-1) α and β, and deficits in proteostasis, characterized by accumulation of polyubiquitinated proteins and other aggregates, are associated with neurodegenerative disease independently and through interactions of the two phenomena. We investigated the influence of IL-1β on ubiquitination via its impact on activation of the E3 ligase parkin by either phosphorylated ubiquitin (P-Ub) or NEDD8. Methods Immunohistochemistry and Proximity Ligation Assay were used to assess colocalization of parkin with P-tau or NEDD8 in hippocampus from Alzheimer patients (AD) and controls. IL-1β effects on PINK1, P-Ub, parkin, P-parkin, and GSK3β—as well as phosphorylation of parkin by GSK3β—were assessed in cell cultures by western immunoblot, using two inhibitors and siRNA knockdown to suppress GSK3β. Computer modeling characterized the binding and the effects of P-Ub and NEDD8 on parkin. IL-1α, IL-1β, and parkin gene expression was assessed by RT-PCR in brains of 2- and 17-month-old PD-APP mice and wild-type littermates. Results IL-1α, IL-1β, and parkin mRNA levels were higher in PD-APP mice compared with wild-type littermates, and IL-1α-laden glia surrounded parkin- and P-tau-laden neurons in human AD. Such neurons showed a nuclear-to-cytoplasmic translocation of NEDD8 that was mimicked in IL-1β-treated primary neuronal cultures. These cultures also showed higher parkin levels and GSK3β-induced parkin phosphorylation; PINK1 levels were suppressed. In silico simulation predicted that binding of either P-Ub or NEDD8 at a singular position on parkin opens the UBL domain, exposing Ser65 for parkin activation. Conclusions The promotion of parkin- and NEDD8-mediated ubiquitination by IL-1β is consistent with an acute neuroprotective role. However, accumulations of P-tau and P-Ub and other elements of proteostasis, such as translocated NEDD8, in AD and in response to IL-1β suggest either over-stimulation or a proteostatic failure that may result from chronic IL-1β elevation, easily envisioned considering its early induction in Down’s syndrome and mild cognitive impairment. The findings further link autophagy and neuroinflammation, two important aspects of AD pathogenesis, which have previously been only loosely related.https://doi.org/10.1186/s12974-019-1669-zIL-1βGSK3βPINK1Alzheimer’sParkinNEDD8
collection DOAJ
language English
format Article
sources DOAJ
author Meenakshisundaram Balasubramaniam
Paul A. Parcon
Chhanda Bose
Ling Liu
Richard A. Jones
Martin R. Farlow
Robert E. Mrak
Steven W. Barger
W. Sue T. Griffin
spellingShingle Meenakshisundaram Balasubramaniam
Paul A. Parcon
Chhanda Bose
Ling Liu
Richard A. Jones
Martin R. Farlow
Robert E. Mrak
Steven W. Barger
W. Sue T. Griffin
Interleukin-1β drives NEDD8 nuclear-to-cytoplasmic translocation, fostering parkin activation via NEDD8 binding to the P-ubiquitin activating site
Journal of Neuroinflammation
IL-1β
GSK3β
PINK1
Alzheimer’s
Parkin
NEDD8
author_facet Meenakshisundaram Balasubramaniam
Paul A. Parcon
Chhanda Bose
Ling Liu
Richard A. Jones
Martin R. Farlow
Robert E. Mrak
Steven W. Barger
W. Sue T. Griffin
author_sort Meenakshisundaram Balasubramaniam
title Interleukin-1β drives NEDD8 nuclear-to-cytoplasmic translocation, fostering parkin activation via NEDD8 binding to the P-ubiquitin activating site
title_short Interleukin-1β drives NEDD8 nuclear-to-cytoplasmic translocation, fostering parkin activation via NEDD8 binding to the P-ubiquitin activating site
title_full Interleukin-1β drives NEDD8 nuclear-to-cytoplasmic translocation, fostering parkin activation via NEDD8 binding to the P-ubiquitin activating site
title_fullStr Interleukin-1β drives NEDD8 nuclear-to-cytoplasmic translocation, fostering parkin activation via NEDD8 binding to the P-ubiquitin activating site
title_full_unstemmed Interleukin-1β drives NEDD8 nuclear-to-cytoplasmic translocation, fostering parkin activation via NEDD8 binding to the P-ubiquitin activating site
title_sort interleukin-1β drives nedd8 nuclear-to-cytoplasmic translocation, fostering parkin activation via nedd8 binding to the p-ubiquitin activating site
publisher BMC
series Journal of Neuroinflammation
issn 1742-2094
publishDate 2019-12-01
description Abstract Background Neuroinflammation, typified by elevated levels of interleukin-1 (IL-1) α and β, and deficits in proteostasis, characterized by accumulation of polyubiquitinated proteins and other aggregates, are associated with neurodegenerative disease independently and through interactions of the two phenomena. We investigated the influence of IL-1β on ubiquitination via its impact on activation of the E3 ligase parkin by either phosphorylated ubiquitin (P-Ub) or NEDD8. Methods Immunohistochemistry and Proximity Ligation Assay were used to assess colocalization of parkin with P-tau or NEDD8 in hippocampus from Alzheimer patients (AD) and controls. IL-1β effects on PINK1, P-Ub, parkin, P-parkin, and GSK3β—as well as phosphorylation of parkin by GSK3β—were assessed in cell cultures by western immunoblot, using two inhibitors and siRNA knockdown to suppress GSK3β. Computer modeling characterized the binding and the effects of P-Ub and NEDD8 on parkin. IL-1α, IL-1β, and parkin gene expression was assessed by RT-PCR in brains of 2- and 17-month-old PD-APP mice and wild-type littermates. Results IL-1α, IL-1β, and parkin mRNA levels were higher in PD-APP mice compared with wild-type littermates, and IL-1α-laden glia surrounded parkin- and P-tau-laden neurons in human AD. Such neurons showed a nuclear-to-cytoplasmic translocation of NEDD8 that was mimicked in IL-1β-treated primary neuronal cultures. These cultures also showed higher parkin levels and GSK3β-induced parkin phosphorylation; PINK1 levels were suppressed. In silico simulation predicted that binding of either P-Ub or NEDD8 at a singular position on parkin opens the UBL domain, exposing Ser65 for parkin activation. Conclusions The promotion of parkin- and NEDD8-mediated ubiquitination by IL-1β is consistent with an acute neuroprotective role. However, accumulations of P-tau and P-Ub and other elements of proteostasis, such as translocated NEDD8, in AD and in response to IL-1β suggest either over-stimulation or a proteostatic failure that may result from chronic IL-1β elevation, easily envisioned considering its early induction in Down’s syndrome and mild cognitive impairment. The findings further link autophagy and neuroinflammation, two important aspects of AD pathogenesis, which have previously been only loosely related.
topic IL-1β
GSK3β
PINK1
Alzheimer’s
Parkin
NEDD8
url https://doi.org/10.1186/s12974-019-1669-z
work_keys_str_mv AT meenakshisundarambalasubramaniam interleukin1bdrivesnedd8nucleartocytoplasmictranslocationfosteringparkinactivationvianedd8bindingtothepubiquitinactivatingsite
AT paulaparcon interleukin1bdrivesnedd8nucleartocytoplasmictranslocationfosteringparkinactivationvianedd8bindingtothepubiquitinactivatingsite
AT chhandabose interleukin1bdrivesnedd8nucleartocytoplasmictranslocationfosteringparkinactivationvianedd8bindingtothepubiquitinactivatingsite
AT lingliu interleukin1bdrivesnedd8nucleartocytoplasmictranslocationfosteringparkinactivationvianedd8bindingtothepubiquitinactivatingsite
AT richardajones interleukin1bdrivesnedd8nucleartocytoplasmictranslocationfosteringparkinactivationvianedd8bindingtothepubiquitinactivatingsite
AT martinrfarlow interleukin1bdrivesnedd8nucleartocytoplasmictranslocationfosteringparkinactivationvianedd8bindingtothepubiquitinactivatingsite
AT robertemrak interleukin1bdrivesnedd8nucleartocytoplasmictranslocationfosteringparkinactivationvianedd8bindingtothepubiquitinactivatingsite
AT stevenwbarger interleukin1bdrivesnedd8nucleartocytoplasmictranslocationfosteringparkinactivationvianedd8bindingtothepubiquitinactivatingsite
AT wsuetgriffin interleukin1bdrivesnedd8nucleartocytoplasmictranslocationfosteringparkinactivationvianedd8bindingtothepubiquitinactivatingsite
_version_ 1724369352476590080