Cargando…

Pharmacological Basis for the Use of Evodiamine in Alzheimer’s Disease: Antioxidation and Antiapoptosis

Evodiamine (Evo), a major alkaloid compound isolated from the dry unripened fruit of Evodia fructus, has a wide range of pharmacological activities. The present study sought to explore the neuroprotective effects of Evo in l-glutamate (l-Glu)-induced apoptosis of HT22 cells, and in a d-galactose and...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Yongfeng, Wang, Jiaqi, Wang, Chunyue, Li, Zhiping, Liu, Xin, Zhang, Jun, Lu, Jiahui, Wang, Di
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5983845/
https://www.ncbi.nlm.nih.gov/pubmed/29883380
http://dx.doi.org/10.3390/ijms19051527
_version_ 1783328513067057152
author Zhang, Yongfeng
Wang, Jiaqi
Wang, Chunyue
Li, Zhiping
Liu, Xin
Zhang, Jun
Lu, Jiahui
Wang, Di
author_facet Zhang, Yongfeng
Wang, Jiaqi
Wang, Chunyue
Li, Zhiping
Liu, Xin
Zhang, Jun
Lu, Jiahui
Wang, Di
author_sort Zhang, Yongfeng
collection PubMed
description Evodiamine (Evo), a major alkaloid compound isolated from the dry unripened fruit of Evodia fructus, has a wide range of pharmacological activities. The present study sought to explore the neuroprotective effects of Evo in l-glutamate (l-Glu)-induced apoptosis of HT22 cells, and in a d-galactose and aluminum trichloride-developed Alzheimer’s disease (AD) mouse model. Evo significantly enhanced cell viability, inhibited the accumulation of reactive oxygen species, ameliorated mitochondrial function, increased the B-cell lymphoma-2 protein content, and inhibited the high expression levels of Bax, Bad, and cleaved-caspase-3 and -8 in l-Glu-induced HT22 cells. Evo also enhanced the phosphorylation activities of protein kinase B and the mammalian target of rapamycin in the l-Glu-induced HT22 cells. In the AD mouse model, Evo reduced the aimless and chaotic movements, reduced the time spent in the central area in the open field test, and decreased the escape latency time in the Morris water maze test. Evo reduced the deposition of amyloid beta 42 (Aβ42) in the brain, and increased the serum level of Aβ42, but showed no significant effects on Aβ40. In addition, six weeks of Evo administration significantly suppressed oxidative stress by modulating the related enzyme levels. In the central cholinergic system of AD mice, Evo significantly increased the serum levels of acetylcholine and choline acetyltransferase and decreased the level of acetylcholinesterase in the serum, hypothalamus, and brain. Our results provide experimental evidence that Evo can serve as a neuroprotective candidate for the prevention and/or treatment of neurodegenerative diseases.
format Online
Article
Text
id pubmed-5983845
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-59838452018-06-05 Pharmacological Basis for the Use of Evodiamine in Alzheimer’s Disease: Antioxidation and Antiapoptosis Zhang, Yongfeng Wang, Jiaqi Wang, Chunyue Li, Zhiping Liu, Xin Zhang, Jun Lu, Jiahui Wang, Di Int J Mol Sci Article Evodiamine (Evo), a major alkaloid compound isolated from the dry unripened fruit of Evodia fructus, has a wide range of pharmacological activities. The present study sought to explore the neuroprotective effects of Evo in l-glutamate (l-Glu)-induced apoptosis of HT22 cells, and in a d-galactose and aluminum trichloride-developed Alzheimer’s disease (AD) mouse model. Evo significantly enhanced cell viability, inhibited the accumulation of reactive oxygen species, ameliorated mitochondrial function, increased the B-cell lymphoma-2 protein content, and inhibited the high expression levels of Bax, Bad, and cleaved-caspase-3 and -8 in l-Glu-induced HT22 cells. Evo also enhanced the phosphorylation activities of protein kinase B and the mammalian target of rapamycin in the l-Glu-induced HT22 cells. In the AD mouse model, Evo reduced the aimless and chaotic movements, reduced the time spent in the central area in the open field test, and decreased the escape latency time in the Morris water maze test. Evo reduced the deposition of amyloid beta 42 (Aβ42) in the brain, and increased the serum level of Aβ42, but showed no significant effects on Aβ40. In addition, six weeks of Evo administration significantly suppressed oxidative stress by modulating the related enzyme levels. In the central cholinergic system of AD mice, Evo significantly increased the serum levels of acetylcholine and choline acetyltransferase and decreased the level of acetylcholinesterase in the serum, hypothalamus, and brain. Our results provide experimental evidence that Evo can serve as a neuroprotective candidate for the prevention and/or treatment of neurodegenerative diseases. MDPI 2018-05-21 /pmc/articles/PMC5983845/ /pubmed/29883380 http://dx.doi.org/10.3390/ijms19051527 Text en © 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Zhang, Yongfeng
Wang, Jiaqi
Wang, Chunyue
Li, Zhiping
Liu, Xin
Zhang, Jun
Lu, Jiahui
Wang, Di
Pharmacological Basis for the Use of Evodiamine in Alzheimer’s Disease: Antioxidation and Antiapoptosis
title Pharmacological Basis for the Use of Evodiamine in Alzheimer’s Disease: Antioxidation and Antiapoptosis
title_full Pharmacological Basis for the Use of Evodiamine in Alzheimer’s Disease: Antioxidation and Antiapoptosis
title_fullStr Pharmacological Basis for the Use of Evodiamine in Alzheimer’s Disease: Antioxidation and Antiapoptosis
title_full_unstemmed Pharmacological Basis for the Use of Evodiamine in Alzheimer’s Disease: Antioxidation and Antiapoptosis
title_short Pharmacological Basis for the Use of Evodiamine in Alzheimer’s Disease: Antioxidation and Antiapoptosis
title_sort pharmacological basis for the use of evodiamine in alzheimer’s disease: antioxidation and antiapoptosis
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5983845/
https://www.ncbi.nlm.nih.gov/pubmed/29883380
http://dx.doi.org/10.3390/ijms19051527
work_keys_str_mv AT zhangyongfeng pharmacologicalbasisfortheuseofevodiamineinalzheimersdiseaseantioxidationandantiapoptosis
AT wangjiaqi pharmacologicalbasisfortheuseofevodiamineinalzheimersdiseaseantioxidationandantiapoptosis
AT wangchunyue pharmacologicalbasisfortheuseofevodiamineinalzheimersdiseaseantioxidationandantiapoptosis
AT lizhiping pharmacologicalbasisfortheuseofevodiamineinalzheimersdiseaseantioxidationandantiapoptosis
AT liuxin pharmacologicalbasisfortheuseofevodiamineinalzheimersdiseaseantioxidationandantiapoptosis
AT zhangjun pharmacologicalbasisfortheuseofevodiamineinalzheimersdiseaseantioxidationandantiapoptosis
AT lujiahui pharmacologicalbasisfortheuseofevodiamineinalzheimersdiseaseantioxidationandantiapoptosis
AT wangdi pharmacologicalbasisfortheuseofevodiamineinalzheimersdiseaseantioxidationandantiapoptosis