PIAS2-mediated blockade of IFN-β signaling: a basis for sporadic Parkinson disease dementia

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

PIAS2-mediated blockade of IFN-β signaling : a basis for sporadic Parkinson disease dementia. / Magalhaes, Joana; Tresse, Emilie; Ejlerskov, Patrick; Hu, Erling; Liu, Yawei; Marin, Andrea; Montalant, Alexia; Satriano, Letizia; Rundsten, Carsten Friis; Carlsen, Eva Maria Meier; Rydbirk, Rasmus; Sharifi-Zarchi, Ali; Andersen, Jesper Bøje; Aznar, Susana; Brudek, Tomasz; Khodosevich, Konstantin; Prinz, Marco; Perrier, Jean François Marie; Sharma, Manu; Gasser, Thomas; Issazadeh-Navikas, Shohreh.

In: Molecular Psychiatry, Vol. 26, 2021, p. 6083–6099.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Magalhaes, J, Tresse, E, Ejlerskov, P, Hu, E, Liu, Y, Marin, A, Montalant, A, Satriano, L, Rundsten, CF, Carlsen, EMM, Rydbirk, R, Sharifi-Zarchi, A, Andersen, JB, Aznar, S, Brudek, T, Khodosevich, K, Prinz, M, Perrier, JFM, Sharma, M, Gasser, T & Issazadeh-Navikas, S 2021, 'PIAS2-mediated blockade of IFN-β signaling: a basis for sporadic Parkinson disease dementia', Molecular Psychiatry, vol. 26, pp. 6083–6099. https://doi.org/10.1038/s41380-021-01207-w

APA

Magalhaes, J., Tresse, E., Ejlerskov, P., Hu, E., Liu, Y., Marin, A., Montalant, A., Satriano, L., Rundsten, C. F., Carlsen, E. M. M., Rydbirk, R., Sharifi-Zarchi, A., Andersen, J. B., Aznar, S., Brudek, T., Khodosevich, K., Prinz, M., Perrier, J. F. M., Sharma, M., ... Issazadeh-Navikas, S. (2021). PIAS2-mediated blockade of IFN-β signaling: a basis for sporadic Parkinson disease dementia. Molecular Psychiatry, 26, 6083–6099. https://doi.org/10.1038/s41380-021-01207-w

Vancouver

Magalhaes J, Tresse E, Ejlerskov P, Hu E, Liu Y, Marin A et al. PIAS2-mediated blockade of IFN-β signaling: a basis for sporadic Parkinson disease dementia. Molecular Psychiatry. 2021;26:6083–6099. https://doi.org/10.1038/s41380-021-01207-w

Author

Magalhaes, Joana ; Tresse, Emilie ; Ejlerskov, Patrick ; Hu, Erling ; Liu, Yawei ; Marin, Andrea ; Montalant, Alexia ; Satriano, Letizia ; Rundsten, Carsten Friis ; Carlsen, Eva Maria Meier ; Rydbirk, Rasmus ; Sharifi-Zarchi, Ali ; Andersen, Jesper Bøje ; Aznar, Susana ; Brudek, Tomasz ; Khodosevich, Konstantin ; Prinz, Marco ; Perrier, Jean François Marie ; Sharma, Manu ; Gasser, Thomas ; Issazadeh-Navikas, Shohreh. / PIAS2-mediated blockade of IFN-β signaling : a basis for sporadic Parkinson disease dementia. In: Molecular Psychiatry. 2021 ; Vol. 26. pp. 6083–6099.

Bibtex

@article{d386990fe2ac44daa12761b65c96e865,
title = "PIAS2-mediated blockade of IFN-β signaling: a basis for sporadic Parkinson disease dementia",
abstract = "Familial Parkinson disease (PD) is associated with rare genetic mutations, but the etiology in most patients with sporadic (s)PD is largely unknown, and the basis for its progression to dementia (sPDD) is poorly characterized. We have identified that loss of IFNβ or IFNAR1, the receptor for IFNα/β, causes pathological and behavioral changes resembling PDD, prompting us to hypothesize that dysregulated genes in IFNβ-IFNAR signaling pathway predispose one to sPD. By transcriptomic analysis, we found defective neuronal IFNβ-IFNAR signaling, including particularly elevated PIAS2 associated with sPDD. With meta-analysis of GWASs, we identified sequence variants in IFNβ-IFNAR-related genes in sPD patients. Furthermore, sPDD patients expressed higher levels of PIAS2 mRNA and protein in neurons. To determine its function in brain, we overexpressed PIAS2 under a neuronal promoter, alone or with human α-synuclein, in the brains of mice, which caused motor and cognitive impairments and correlated with intraneuronal phosphorylated (p)α-synuclein accumulation and dopaminergic neuron loss. Ectopic expression of neuronal PIAS2 blocked mitophagy, increased the accumulation of senescent mitochondrial and oxidative stress, as evidenced by excessive oxDJ1 and 8OHdG, by inactivating ERK1/2-P53 signaling. Conversely, PIAS2 knockdown rescued the clinicopathological manifestations of PDD in Ifnb–/– mice on restoring mitochondrial homeostasis, oxidative stress, and pERK1/2-pP53 signaling. The regulation of JAK-STAT2-PIAS2 signaling was crucial for neurite outgrowth and neuronal survival and excitability and thus might prevent cognitive impairments. Our findings provide insights into the progression of sPD and dementia and have implications for new therapeutic approaches.",
author = "Joana Magalhaes and Emilie Tresse and Patrick Ejlerskov and Erling Hu and Yawei Liu and Andrea Marin and Alexia Montalant and Letizia Satriano and Rundsten, {Carsten Friis} and Carlsen, {Eva Maria Meier} and Rasmus Rydbirk and Ali Sharifi-Zarchi and Andersen, {Jesper B{\o}je} and Susana Aznar and Tomasz Brudek and Konstantin Khodosevich and Marco Prinz and Perrier, {Jean Fran{\c c}ois Marie} and Manu Sharma and Thomas Gasser and Shohreh Issazadeh-Navikas",
year = "2021",
doi = "10.1038/s41380-021-01207-w",
language = "English",
volume = "26",
pages = "6083–6099",
journal = "Molecular Psychiatry",
issn = "1359-4184",
publisher = "nature publishing group",

}

RIS

TY - JOUR

T1 - PIAS2-mediated blockade of IFN-β signaling

T2 - a basis for sporadic Parkinson disease dementia

AU - Magalhaes, Joana

AU - Tresse, Emilie

AU - Ejlerskov, Patrick

AU - Hu, Erling

AU - Liu, Yawei

AU - Marin, Andrea

AU - Montalant, Alexia

AU - Satriano, Letizia

AU - Rundsten, Carsten Friis

AU - Carlsen, Eva Maria Meier

AU - Rydbirk, Rasmus

AU - Sharifi-Zarchi, Ali

AU - Andersen, Jesper Bøje

AU - Aznar, Susana

AU - Brudek, Tomasz

AU - Khodosevich, Konstantin

AU - Prinz, Marco

AU - Perrier, Jean François Marie

AU - Sharma, Manu

AU - Gasser, Thomas

AU - Issazadeh-Navikas, Shohreh

PY - 2021

Y1 - 2021

N2 - Familial Parkinson disease (PD) is associated with rare genetic mutations, but the etiology in most patients with sporadic (s)PD is largely unknown, and the basis for its progression to dementia (sPDD) is poorly characterized. We have identified that loss of IFNβ or IFNAR1, the receptor for IFNα/β, causes pathological and behavioral changes resembling PDD, prompting us to hypothesize that dysregulated genes in IFNβ-IFNAR signaling pathway predispose one to sPD. By transcriptomic analysis, we found defective neuronal IFNβ-IFNAR signaling, including particularly elevated PIAS2 associated with sPDD. With meta-analysis of GWASs, we identified sequence variants in IFNβ-IFNAR-related genes in sPD patients. Furthermore, sPDD patients expressed higher levels of PIAS2 mRNA and protein in neurons. To determine its function in brain, we overexpressed PIAS2 under a neuronal promoter, alone or with human α-synuclein, in the brains of mice, which caused motor and cognitive impairments and correlated with intraneuronal phosphorylated (p)α-synuclein accumulation and dopaminergic neuron loss. Ectopic expression of neuronal PIAS2 blocked mitophagy, increased the accumulation of senescent mitochondrial and oxidative stress, as evidenced by excessive oxDJ1 and 8OHdG, by inactivating ERK1/2-P53 signaling. Conversely, PIAS2 knockdown rescued the clinicopathological manifestations of PDD in Ifnb–/– mice on restoring mitochondrial homeostasis, oxidative stress, and pERK1/2-pP53 signaling. The regulation of JAK-STAT2-PIAS2 signaling was crucial for neurite outgrowth and neuronal survival and excitability and thus might prevent cognitive impairments. Our findings provide insights into the progression of sPD and dementia and have implications for new therapeutic approaches.

AB - Familial Parkinson disease (PD) is associated with rare genetic mutations, but the etiology in most patients with sporadic (s)PD is largely unknown, and the basis for its progression to dementia (sPDD) is poorly characterized. We have identified that loss of IFNβ or IFNAR1, the receptor for IFNα/β, causes pathological and behavioral changes resembling PDD, prompting us to hypothesize that dysregulated genes in IFNβ-IFNAR signaling pathway predispose one to sPD. By transcriptomic analysis, we found defective neuronal IFNβ-IFNAR signaling, including particularly elevated PIAS2 associated with sPDD. With meta-analysis of GWASs, we identified sequence variants in IFNβ-IFNAR-related genes in sPD patients. Furthermore, sPDD patients expressed higher levels of PIAS2 mRNA and protein in neurons. To determine its function in brain, we overexpressed PIAS2 under a neuronal promoter, alone or with human α-synuclein, in the brains of mice, which caused motor and cognitive impairments and correlated with intraneuronal phosphorylated (p)α-synuclein accumulation and dopaminergic neuron loss. Ectopic expression of neuronal PIAS2 blocked mitophagy, increased the accumulation of senescent mitochondrial and oxidative stress, as evidenced by excessive oxDJ1 and 8OHdG, by inactivating ERK1/2-P53 signaling. Conversely, PIAS2 knockdown rescued the clinicopathological manifestations of PDD in Ifnb–/– mice on restoring mitochondrial homeostasis, oxidative stress, and pERK1/2-pP53 signaling. The regulation of JAK-STAT2-PIAS2 signaling was crucial for neurite outgrowth and neuronal survival and excitability and thus might prevent cognitive impairments. Our findings provide insights into the progression of sPD and dementia and have implications for new therapeutic approaches.

U2 - 10.1038/s41380-021-01207-w

DO - 10.1038/s41380-021-01207-w

M3 - Journal article

C2 - 34234281

VL - 26

SP - 6083

EP - 6099

JO - Molecular Psychiatry

JF - Molecular Psychiatry

SN - 1359-4184

ER -

ID: 282552660