Synthetic Peucedanocoumarin IV Prevents α-Synuclein Neurotoxicity in an Animal Model of Parkinson’s Disease

Pathological protein inclusion formation and propagation are the main causes of neuronal dysfunction in diverse neurodegenerative diseases; therefore, current disease-modifying therapeutic strategies have targeted this disease protein aggregation process. Recently, we reported that peucedanocoumarin...

Full description

Bibliographic Details
Main Authors: Heejeong Kim, Han-Joo Maeng, Ji Hun Kim, Jin-Ha Yoon, Yohan Oh, Seung-Mann Paek, Yunjong Lee
Format: Article
Language:English
Published: MDPI AG 2022-08-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/23/15/8618
_version_ 1797432863645237248
author Heejeong Kim
Han-Joo Maeng
Ji Hun Kim
Jin-Ha Yoon
Yohan Oh
Seung-Mann Paek
Yunjong Lee
author_facet Heejeong Kim
Han-Joo Maeng
Ji Hun Kim
Jin-Ha Yoon
Yohan Oh
Seung-Mann Paek
Yunjong Lee
author_sort Heejeong Kim
collection DOAJ
description Pathological protein inclusion formation and propagation are the main causes of neuronal dysfunction in diverse neurodegenerative diseases; therefore, current disease-modifying therapeutic strategies have targeted this disease protein aggregation process. Recently, we reported that peucedanocoumarin III (PCiii) is a promising therapeutic compound with the ability to disaggregate α-synuclein inclusion and protect dopaminergic neurons in Parkinson’s disease (PD). Here, we found that <i>trans</i>-4′-acetyl-3′-tigloylkhellactone (racemic peucedanocoumarin IV [PCiv]), a structural isomer of PCiii with a higher synthetic yield presented a strong anti-aggregate activity to a degree comparable to that of PCiii. PCiv retained effective inhibitory function against β-sheet aggregate-mimic β23 cytotoxicities and potently prevented α-synucleinopathy in α-synuclein preformed fibril (PFF)-treated mice cortical neurons. In detailed pharmacokinetic profiling of PCiv, oral administration of PCiv in rats exhibited an approximately 97-min half-life and 10% bioavailability. Moreover, tissue distribution analysis revealed favorable profiles of brain penetration with a 6.4 brain-to-plasma concentration ratio. The therapeutic efficacy of PCiv was further evaluated in a sporadic PD mouse model with a combinatorial co-injection of α-synuclein preformed fibril and recombinant adeno-associated virus expressing α-synuclein. Motor dysfunctions induced in this combinatorial α-synucleinopathy PD mouse model was almost completely rescued by PCiv diet administration, and this therapeutic effect is consistent with the marked prevention of dopaminergic neuron loss and suppression of α-synuclein aggregation. Taken together, our translational study suggests that PCiv is advantageous as a therapeutic agent for neurodegenerative diseases, especially with its good synthetic yield, high brain distribution, and anti-aggregate activity. PCiv may be useful in the management of α-synuclein inclusion formation and propagation at different stages of PD.
first_indexed 2024-03-09T10:07:49Z
format Article
id doaj.art-c6de75b53a784716861a368f03f0f427
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-09T10:07:49Z
publishDate 2022-08-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-c6de75b53a784716861a368f03f0f4272023-12-01T22:58:17ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672022-08-012315861810.3390/ijms23158618Synthetic Peucedanocoumarin IV Prevents α-Synuclein Neurotoxicity in an Animal Model of Parkinson’s DiseaseHeejeong Kim0Han-Joo Maeng1Ji Hun Kim2Jin-Ha Yoon3Yohan Oh4Seung-Mann Paek5Yunjong Lee6Department of Pharmacology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 16419, KoreaCollege of Pharmacy, Gachon University, Incheon 21936, KoreaDepartment of Pharmacology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 16419, KoreaCollege of Pharmacy, Gachon University, Incheon 21936, KoreaDepartment of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, KoreaCollege of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju Daero 501, Jinju 52828, KoreaDepartment of Pharmacology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 16419, KoreaPathological protein inclusion formation and propagation are the main causes of neuronal dysfunction in diverse neurodegenerative diseases; therefore, current disease-modifying therapeutic strategies have targeted this disease protein aggregation process. Recently, we reported that peucedanocoumarin III (PCiii) is a promising therapeutic compound with the ability to disaggregate α-synuclein inclusion and protect dopaminergic neurons in Parkinson’s disease (PD). Here, we found that <i>trans</i>-4′-acetyl-3′-tigloylkhellactone (racemic peucedanocoumarin IV [PCiv]), a structural isomer of PCiii with a higher synthetic yield presented a strong anti-aggregate activity to a degree comparable to that of PCiii. PCiv retained effective inhibitory function against β-sheet aggregate-mimic β23 cytotoxicities and potently prevented α-synucleinopathy in α-synuclein preformed fibril (PFF)-treated mice cortical neurons. In detailed pharmacokinetic profiling of PCiv, oral administration of PCiv in rats exhibited an approximately 97-min half-life and 10% bioavailability. Moreover, tissue distribution analysis revealed favorable profiles of brain penetration with a 6.4 brain-to-plasma concentration ratio. The therapeutic efficacy of PCiv was further evaluated in a sporadic PD mouse model with a combinatorial co-injection of α-synuclein preformed fibril and recombinant adeno-associated virus expressing α-synuclein. Motor dysfunctions induced in this combinatorial α-synucleinopathy PD mouse model was almost completely rescued by PCiv diet administration, and this therapeutic effect is consistent with the marked prevention of dopaminergic neuron loss and suppression of α-synuclein aggregation. Taken together, our translational study suggests that PCiv is advantageous as a therapeutic agent for neurodegenerative diseases, especially with its good synthetic yield, high brain distribution, and anti-aggregate activity. PCiv may be useful in the management of α-synuclein inclusion formation and propagation at different stages of PD.https://www.mdpi.com/1422-0067/23/15/8618peucedanocoumarin IVorganic synthesisParkinson’s diseasepharmacokineticsα-synuclein preformed fibrildopaminergic cell loss
spellingShingle Heejeong Kim
Han-Joo Maeng
Ji Hun Kim
Jin-Ha Yoon
Yohan Oh
Seung-Mann Paek
Yunjong Lee
Synthetic Peucedanocoumarin IV Prevents α-Synuclein Neurotoxicity in an Animal Model of Parkinson’s Disease
International Journal of Molecular Sciences
peucedanocoumarin IV
organic synthesis
Parkinson’s disease
pharmacokinetics
α-synuclein preformed fibril
dopaminergic cell loss
title Synthetic Peucedanocoumarin IV Prevents α-Synuclein Neurotoxicity in an Animal Model of Parkinson’s Disease
title_full Synthetic Peucedanocoumarin IV Prevents α-Synuclein Neurotoxicity in an Animal Model of Parkinson’s Disease
title_fullStr Synthetic Peucedanocoumarin IV Prevents α-Synuclein Neurotoxicity in an Animal Model of Parkinson’s Disease
title_full_unstemmed Synthetic Peucedanocoumarin IV Prevents α-Synuclein Neurotoxicity in an Animal Model of Parkinson’s Disease
title_short Synthetic Peucedanocoumarin IV Prevents α-Synuclein Neurotoxicity in an Animal Model of Parkinson’s Disease
title_sort synthetic peucedanocoumarin iv prevents α synuclein neurotoxicity in an animal model of parkinson s disease
topic peucedanocoumarin IV
organic synthesis
Parkinson’s disease
pharmacokinetics
α-synuclein preformed fibril
dopaminergic cell loss
url https://www.mdpi.com/1422-0067/23/15/8618
work_keys_str_mv AT heejeongkim syntheticpeucedanocoumarinivpreventsasynucleinneurotoxicityinananimalmodelofparkinsonsdisease
AT hanjoomaeng syntheticpeucedanocoumarinivpreventsasynucleinneurotoxicityinananimalmodelofparkinsonsdisease
AT jihunkim syntheticpeucedanocoumarinivpreventsasynucleinneurotoxicityinananimalmodelofparkinsonsdisease
AT jinhayoon syntheticpeucedanocoumarinivpreventsasynucleinneurotoxicityinananimalmodelofparkinsonsdisease
AT yohanoh syntheticpeucedanocoumarinivpreventsasynucleinneurotoxicityinananimalmodelofparkinsonsdisease
AT seungmannpaek syntheticpeucedanocoumarinivpreventsasynucleinneurotoxicityinananimalmodelofparkinsonsdisease
AT yunjonglee syntheticpeucedanocoumarinivpreventsasynucleinneurotoxicityinananimalmodelofparkinsonsdisease