1
|
Intharuksa A, Kuljarusnont S, Sasaki Y, Tungmunnithum D. Flavonoids and Other Polyphenols: Bioactive Molecules from Traditional Medicine Recipes/Medicinal Plants and Their Potential for Phytopharmaceutical and Medical Application. Molecules 2024; 29:5760. [PMID: 39683916 DOI: 10.3390/molecules29235760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Currently, natural bioactive ingredients and/or raw materials are of significant interest to scientists around the world. Flavonoids and other polyphenols are a major group of phytochemicals that have been researched and noted as bioactive molecules. They offer several pharmacological and medical benefits. This current review aims to (1) illustrate their benefits for human health, such as antioxidant, anti-aging, anti-cancer, anti-inflammatory, anti-microbial, cardioprotective, neuroprotective, and UV-protective effects, and also (2) to perform a quality evaluation of traditional medicines for future application. Consequently, keywords were searched on Scopus, Google Scholar, and PubMed so as to search for related publications. Then, those publications were carefully checked in order to find current and non-redundant studies that matched the objective of this review. According to this review, researchers worldwide are very interested in discovering the potential of flavonoids and other polyphenols, used in traditional medicines and taken from medicinal plants, in relation to medical and pharmaceutical applications. Many studies focus on the health benefits of flavonoids and other polyphenols have been tested using in silico, in vitro, and in vivo models. However, few studies have been carried out using clinical trials that have trustworthy subject sizes and are in accordance with clinical practice guidelines. Additionally, interesting research directions and perspectives for future studies are highlighted in this work.
Collapse
Affiliation(s)
- Aekkhaluck Intharuksa
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sompop Kuljarusnont
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Yohei Sasaki
- Division of Pharmaceutical Sciences, Graduate School of Medical Plant Sciences, Kanazawa University, Kanazawa 920-1192, Japan
| | - Duangjai Tungmunnithum
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Le Studium Institute for Advanced Studies, 1 Rue Dupanloup, 45000 Orléans, France
| |
Collapse
|
2
|
Youn K, Jun M. Determination of Potential Lead Compound from Magnolia officinalis for Alzheimer's Disease through Pharmacokinetic Prediction, Molecular Docking, Dynamic Simulation, and Experimental Validation. Int J Mol Sci 2024; 25:10507. [PMID: 39408835 PMCID: PMC11477134 DOI: 10.3390/ijms251910507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Amyloid β protein (Aβ) deposition has been implicated as the molecular driver of Alzheimer's disease (AD) progression. The modulation of the formation of abnormal aggregates and their post-translational modification is strongly suggested as the most effective approach to anti-AD. Beta-site APP-cleaving enzyme 1 (BACE1) acts upstream in amyloidogenic processing to generate Aβ, which rapidly aggregates alone or in combination with acetylcholinesterase (AChE) to form fibrils. Accumulated Aβ promotes BACE1 activation via glycogen synthase kinase-3β (GSK-3β) and is post-translationally modified by glutaminyl cyclase (QC), resulting in increased neurotoxicity. A novel multi-target inhibitor as a potential AD agent was identified using an in silico approach and experimental validation. Magnolia officinalis, which showed the best anti-AD activity in our preliminary study, was subjected to analysis, and 82 compounds were studied. Among 23 compounds with drug-likeness, blood-brain barrier penetration, and safety, honokiol emerged as a lead structure for the inhibition of BACE1, AChE, QC, and GSK-3β in docking and molecular dynamics (MD) simulations. Furthermore, honokiol was found to be an excellent multi-target inhibitor of these enzymes with an IC50 of 6-90 μM, even when compared to other natural single-target inhibitors. Taken together, the present study is the first to demonstrate that honokiol acts as a multiple enzyme inhibitor with an excellent pharmacokinetic and safety profile which may provide inhibitory effects in broad-range areas including the overproduction, aggregation, and post-translational modification of Aβ. It also provides insight into novel structural features for the design and discovery of multi-target inhibitors for anti-AD.
Collapse
Affiliation(s)
- Kumju Youn
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Republic of Korea;
- Center for Food & Bio Innovation, Dong-A University, Busan 49315, Republic of Korea
| | - Mira Jun
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Republic of Korea;
- Center for Food & Bio Innovation, Dong-A University, Busan 49315, Republic of Korea
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| |
Collapse
|
3
|
Li N, Liang Y, Zhang L, Xu C, Wang L. Neolignans in Magnolia officinalis as natural anti-Alzheimer's disease agents: A systematic review. Ageing Res Rev 2024; 99:102398. [PMID: 38955265 DOI: 10.1016/j.arr.2024.102398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Magnolia officinalis, a traditional herbal medicine widely used in clinical practice, exerts antibacterial, anti-tumor, anti-inflammatory, antioxidant, and anti-aging activities. Neolignans are the main active ingredients of M. officinalis and exert a wide range of pharmacological effects, including anti-Alzheimer's disease (AD) activity. OBJECTIVE To summarize the published data on the therapeutic effect and mechanism of neolignans on AD in vivo and in vitro. METHODS PubMed, Web of Science, Google Scholar, and Scopus were systematically reviewed (up to March 1, 2024) for pre-clinical studies. RESULTS M. officinalis-derived neolignans (honokiol, magnolol, 4-O-methylhonokiol, and obovatol) alleviated behavioral abnormalities, including learning and cognitive impairments, in AD animal models. Mechanistically, neolignans inhibited Aβ generation or aggregation, neuroinflammation, and acetylcholinesterase activity; promoted microglial phagocytosis and anti-oxidative stress; alleviated mitochondrial dysfunction and energy metabolism, as well as anti-cholinergic deficiency; and regulated intestinal flora. Furthermore, neolignans may achieve neuroprotection by regulating different molecular pathways, including the NF-κB, ERK, AMPK/mTOR/ULK1, and cAMP/PKA/CREB pathways. CONCLUSIONS Neolignans exert anti-AD effects through multiple mechanisms and pathways. However, the exact targets, pharmacokinetics, safety, and clinical efficacy in patients with AD need further investigation in multi-center clinical case-control studies.
Collapse
Affiliation(s)
- Na Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110000, PR China.
| | - Yuanyuan Liang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110000, PR China.
| | - Lijuan Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110000, PR China.
| | - Changlu Xu
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110000, PR China.
| | - Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110000, PR China.
| |
Collapse
|
4
|
Arnold RE, Saska J, Mesquita-Ribeiro R, Dajas-Bailador F, Taylor L, Lewis W, Argent S, Shao H, Houk KN, Denton RM. Total synthesis, biological evaluation and biosynthetic re-evaluation of Illicium-derived neolignans. Chem Sci 2024; 15:11783-11793. [PMID: 39092111 PMCID: PMC11290413 DOI: 10.1039/d4sc03232b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/13/2024] [Indexed: 08/04/2024] Open
Abstract
We report the first total syntheses of simonsol F (3), simonsinol (5), fargenin (4), and macranthol (6) in addition to syntheses of simonsol C (2), simonsol G (1), and honokiol (14). The syntheses are based upon a phosphonium ylide-mediated cascade reaction and upon natural product isomerization reactions which proceed through Cope rearrangements of putative biosynthetic dienone intermediates. As a corollary of the natural product isomerization reactions, we propose an alternative biosynthesis of honokiol (14), simonsinol (5), and macranthol (6) which unites the natural products in this family under a single common precursor, chavicol (7). Finally, we demonstrate that simonsol C (2) and simonsol F (3) promote axonal growth in primary mouse cortical neurons.
Collapse
Affiliation(s)
- Robert E Arnold
- The GlaxoSmithKline Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham Jubilee Campus Triumph Road Nottingham NG7 2TU UK
| | - Jan Saska
- The GlaxoSmithKline Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham Jubilee Campus Triumph Road Nottingham NG7 2TU UK
| | | | | | - Laurence Taylor
- The GlaxoSmithKline Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham Jubilee Campus Triumph Road Nottingham NG7 2TU UK
| | - William Lewis
- The GlaxoSmithKline Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham Jubilee Campus Triumph Road Nottingham NG7 2TU UK
| | - Stephen Argent
- The GlaxoSmithKline Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham Jubilee Campus Triumph Road Nottingham NG7 2TU UK
| | - Huiling Shao
- University of California, Department of Chemistry and Biochemistry 607 Charles E. Young Drive East, Box 951569 Los Angeles CA 90095-1569 UK
| | - Kendall N Houk
- University of California, Department of Chemistry and Biochemistry 607 Charles E. Young Drive East, Box 951569 Los Angeles CA 90095-1569 UK
| | - Ross M Denton
- The GlaxoSmithKline Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham Jubilee Campus Triumph Road Nottingham NG7 2TU UK
| |
Collapse
|
5
|
Wang X, Cheng D, Liu L, Yu H, Wang M. Magnolol ameliorates fumonisin B 1-induced oxidative damage and lipid metabolism dysfunction in astrocyte-like C6 cells. CHEMOSPHERE 2024; 359:142300. [PMID: 38729444 DOI: 10.1016/j.chemosphere.2024.142300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/12/2024]
Abstract
The neurotoxicity of fumonisin B1 (FB1), a commonly detected mycotoxin in crops and the environment, has attracted considerable attention in recent years. However, no effective method for eliminating FB1 completely exists due to the thermal stability and water solubility of this mycotoxin. Magnolol (MAG) is a neolignane with antioxidative and neuroprotective effects. It has been applied in neurotoxicity treatment. However, the application of MAG to attenuate FB1-induced toxicity has not been reported. This study explored the protective mechanism of MAG against FB1-induced damage in C6 cells through antioxidant and lipid metabolism modulation. Results showed that exposure to 15 μM FB1 caused oxidative stress by changing the levels of malondialdehyde, reactive oxygen species, total superoxide dismutase, catalase, and total glutathione. These changes were reversed by MAG addition, especially at the concentration of 80 μM. The protective effects of MAG were further confirmed by the reduction in the phosphorylation levels of proteins in the MAPK signaling pathway. Lipidomics analysis identified 263 lipids, which belong to 24 lipid classes. Among all of the identified lipids, triglycerides (TGs), diglycerides (DGs), phosphatidylcholines (PCs), wax monoesters (WEs), Cers, and phosphatidylethanolamines (PEs) were major categories. Moreover, nine categories of lipids showed the opposite change trend in the FB1 exposure and MAG 80 groups. A further investigation of the 34 co-occurring differential lipids with remarkable changes (P value < 0.05 and VIP value > 1) in the control, FB1 exposure, and MAG 80 groups was performed. Therein, nine lipids (PCs, LPCs, and SM) were screened out as potential biomarkers to reveal the cytoprotective effects of MAG. This work is the first to investigate the rescue mechanism of MAG in FB1-induced cytotoxicity. The obtained results may expand the application of MAG to alleviate the toxicity of mycotoxins.
Collapse
Affiliation(s)
- Xinlu Wang
- Institute of Quality Standard and Testing Technology, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Dai Cheng
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Lin Liu
- Institute of Quality Standard and Testing Technology, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Haiqi Yu
- Institute of Quality Standard and Testing Technology, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Meng Wang
- Institute of Quality Standard and Testing Technology, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China.
| |
Collapse
|
6
|
Szałabska-Rąpała K, Zych M, Borymska W, Londzin P, Dudek S, Kaczmarczyk-Żebrowska I. Beneficial effect of honokiol and magnolol on polyol pathway and oxidative stress parameters in the testes of diabetic rats. Biomed Pharmacother 2024; 172:116265. [PMID: 38364735 DOI: 10.1016/j.biopha.2024.116265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/18/2024] Open
Abstract
In diabetes hyperglycemia, excessive production of free radicals and present oxidative stress lead to many complications in the body, including male reproductive system disorders. To prevent the development of diabetic complications in the testes resulting from them, it seems beneficial to include compounds considered as natural antioxidants. Honokiol and magnolol are neolignans obtained from magnolia bark, which possess proven antioxidant properties. The aim of this study was to evaluate the effect of honokiol and magnolol on the parameters of oxidative stress, polyol pathway and glycation products in the testes as well as on selected biochemical parameters in the blood serum of rats with type 2 diabetes. The study was conducted on mature male Wistar rats with high fat diet and streptozotocin-induced type 2 diabetes. Neolignans-treated rats received honokiol or magnolol orally at the doses of 5 or 25 mg/kg, respectively, for 4 weeks. Parameters related to glucose and lipid homeostasis, basic serological parameters and sex hormones level in the serum as well as polyol pathway parameters, antioxidant enzyme activity, endogenous antioxidants level, sumaric parameters for oxidative stress and oxidative damage in the testes were estimated. Oral administration of honokiol and magnolol turned out to be beneficial in combating the effects of oxidative stess in the testes, but showed no favorable effects on serum biochemical parameters. Additionally, magnolol compared to honokiol revealed more advantageous impact indicating the reversal of the effects of diabetic complications in the male reproductive system and counteracted oxidative stress damages and polyol pathway disorders in the testes.
Collapse
Affiliation(s)
- Katarzyna Szałabska-Rąpała
- Doctoral School of the Medical University of Silesia in Katowice, Discipline of Pharmaceutical Sciences, Department of Pharmacognosy and Phytochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, Sosnowiec 41-200, Poland.
| | - Maria Zych
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, Sosnowiec 41-200, Poland
| | - Weronika Borymska
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, Sosnowiec 41-200, Poland
| | - Piotr Londzin
- Department of Pharmacology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, Sosnowiec 41-200, Poland
| | - Sławomir Dudek
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, Sosnowiec 41-200, Poland
| | - Ilona Kaczmarczyk-Żebrowska
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, Sosnowiec 41-200, Poland
| |
Collapse
|
7
|
Yu J, Gao X, Zhang L, Shi H, Yan Y, Han Y, Wu C, Liu Y, Fang M, Huang C, Fan S. Magnolol extends lifespan and improves age-related neurodegeneration in Caenorhabditis elegans via increase of stress resistance. Sci Rep 2024; 14:3158. [PMID: 38326350 PMCID: PMC10850488 DOI: 10.1038/s41598-024-53374-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/31/2024] [Indexed: 02/09/2024] Open
Abstract
Magnolol is a naturally occurring polyphenolic compound in many edible plants, which has various biological effects including anti-aging and alleviating neurodegenerative diseases. However, the underlying mechanism on longevity is uncertain. In this study, we investigated the effect of magnolol on the lifespan of Caenorhabditis elegans and explored the mechanism. The results showed that magnolol treatment significantly extended the lifespan of nematode and alleviated senescence-related decline in the nematode model. Meanwhile, magnolol enhanced stress resistance to heat shock, hydrogen peroxide (H2O2), mercuric potassium chloride (MeHgCl) and paraquat (PQ) in nematode. In addition, magnolol reduced reactive oxygen species and malondialdehyde (MDA) levels, and increased superoxide dismutase and catalase (CAT) activities in nematodes. Magnolol also up-regulated gene expression of sod-3, hsp16.2, ctl-3, daf-16, skn-1, hsf-1, sir2.1, etc., down-regulated gene expression of daf-2, and promoted intranuclear translocation of daf-16 in nematodes. The lifespan-extending effect of magnolol were reversed in insulin/IGF signaling (IIS) pathway-related mutant lines, including daf-2, age-1, daf-16, skn-1, hsf-1 and sir-2.1, suggesting that IIS signaling is involved in the modulation of longevity by magnolol. Furthermore, magnolol improved the age-related neurodegeneration in PD and AD C. elegans models. These results indicate that magnolol may enhance lifespan and health span through IIS and sir-2.1 pathways. Thus, the current findings implicate magnolol as a potential candidate to ameliorate the symptoms of aging.
Collapse
Affiliation(s)
- Jing Yu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoyan Gao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lijun Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hang Shi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yingxuan Yan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yongli Han
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chengyuan Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ying Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Minglv Fang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
8
|
Fukuyama Y, Kubo M, Harada K. Neurotrophic Natural Products. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 2024; 123:1-473. [PMID: 38340248 DOI: 10.1007/978-3-031-42422-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Neurotrophins (NGF, BDNF, NT3, NT4) can decrease cell death, induce differentiation, as well as sustain the structure and function of neurons, which make them promising therapeutic agents for the treatment of neurodegenerative disorders. However, neurotrophins have not been very effective in clinical trials mostly because they cannot pass through the blood-brain barrier owing to being high-molecular-weight proteins. Thus, neurotrophin-mimic small molecules, which stimulate the synthesis of endogenous neurotrophins or enhance neurotrophic actions, may serve as promising alternatives to neurotrophins. Small-molecular-weight natural products, which have been used in dietary functional foods or in traditional medicines over the course of human history, have a great potential for the development of new therapeutic agents against neurodegenerative diseases such as Alzheimer's disease. In this contribution, a variety of natural products possessing neurotrophic properties such as neurogenesis, neurite outgrowth promotion (neuritogenesis), and neuroprotection are described, and a focus is made on the chemistry and biology of several neurotrophic natural products.
Collapse
Affiliation(s)
- Yoshiyasu Fukuyama
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan.
| | - Miwa Kubo
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan
| | - Kenichi Harada
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan
| |
Collapse
|
9
|
Faysal M, Khan J, Zehravi M, Nath N, Singh LP, Kakkar S, Perusomula R, Khan PA, Nainu F, Asiri M, Khan SL, Das R, Emran TB, Wilairatana P. Neuropharmacological potential of honokiol and its derivatives from Chinese herb Magnolia species: understandings from therapeutic viewpoint. Chin Med 2023; 18:154. [PMID: 38001538 PMCID: PMC10668527 DOI: 10.1186/s13020-023-00846-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/30/2023] [Indexed: 11/26/2023] Open
Abstract
Honokiol is a neolignan biphenol found in aerial parts of the Magnolia plant species. The Magnolia plant species traditionally belong to China and have been used for centuries to treat many pathological conditions. Honokiol mitigates the severity of several pathological conditions and has the potential to work as an anti-inflammatory, anti-angiogenic, anticancer, antioxidant, and neurotherapeutic agent. It has a long history of being employed in the healthcare practices of Southeast Asia, but in recent years, a greater scope of research has been conducted on it. Plenty of experimental evidence suggests it could be beneficial as a neuroprotective bioactive molecule. Honokiol has several pharmacological effects, leading to its exploration as a potential therapy for neurological diseases (NDs), including Alzheimer's disease (AD), Parkinson's disease (PD), cerebral ischemia, anxiety, depression, spinal cord injury, and so on. So, based on the previous experimentation reports, our goal is to discuss the neuroprotective properties of honokiol. Besides, honokiol derivatives have been highlighted recently as possible therapeutic options for NDs. So, this review focuses on honokiol's neurotherapeutic actions and toxicological profile to determine their safety and potential use in neurotherapeutics.
Collapse
Affiliation(s)
- Md Faysal
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Jishan Khan
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chittagong, 4318, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, 51418, Buraydah, Saudi Arabia.
| | - Nikhil Nath
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chittagong, 4318, Bangladesh
| | - Laliteshwar Pratap Singh
- Department of Pharmaceutical Chemistry, Narayan Institute of Pharmacy, Gopal Narayan Singh University, Jamuhar, Sasaram, (Rohtas), Bihar, 821305, India
| | - Saloni Kakkar
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Rajashekar Perusomula
- Cognitive Science Research Initiative Lab, Vishnu Institute of Pharmaceutical Education & Research, Narsapur, India
| | - Pathan Amanulla Khan
- Department of Pharmacy Practice, Anwar Ul Uloom College of Pharmacy, New Mallepally, Hyderabad, India
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Mohammed Asiri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Sharuk L Khan
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa, Maharashtra, 413520, India
| | - Rajib Das
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, USA.
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
10
|
Khatoon F, Ali S, Kumar V, Elasbali AM, Alhassan HH, Alharethi SH, Islam A, Hassan MI. Pharmacological features, health benefits and clinical implications of honokiol. J Biomol Struct Dyn 2023; 41:7511-7533. [PMID: 36093963 DOI: 10.1080/07391102.2022.2120541] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/29/2022] [Indexed: 10/14/2022]
Abstract
Honokiol (HNK) is a natural polyphenolic compound extracted from the bark and leaves of Magnolia grandiflora. It has been traditionally used as a medicinal compound to treat inflammatory diseases. HNK possesses numerous health benefits with a minimal level of toxicity. It can cross the blood-brain barrier and blood-cerebrospinal fluid, thus having significant bioavailability in the neurological tissues. HNK is a promising bioactive compound possesses neuroprotective, antimicrobial, anti-tumorigenic, anti-spasmodic, antidepressant, analgesic, and antithrombotic features . HNK can prevent the growth of several cancer types and haematological malignancies. Recent studies suggested its role in COVID-19 therapy. It binds effectively with several molecular targets, including apoptotic factors, chemokines, transcription factors, cell surface adhesion molecules, and kinases. HNK has excellent pharmacological features and a wide range of chemotherapeutic effects, and thus, researchers have increased interest in improving the therapeutic implications of HNK to the clinic as a novel agent. This review focused on the therapeutic implications of HNK, highlighting clinical and pharmacological features and the underlying mechanism of action.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fatima Khatoon
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| | - Sabeeha Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Vijay Kumar
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, India
| | - Abdelbaset Mohamed Elasbali
- Department of Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Saudi Arabia
| | - Hassan H Alhassan
- Department of Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Saudi Arabia
| | - Salem Hussain Alharethi
- Department of Biological Science, College of Arts and Science, Najran University, Najran, Saudia Arabia
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
11
|
Fagen SJ, Burgess JD, Lim MJ, Amerna D, Kaya ZB, Faroqi AH, Perisetla P, DeMeo NN, Stojkovska I, Quiriconi DJ, Mazzulli JR, Delenclos M, Boschen SL, McLean PJ. Honokiol decreases alpha-synuclein mRNA levels and reveals novel targets for modulating alpha-synuclein expression. Front Aging Neurosci 2023; 15:1179086. [PMID: 37637959 PMCID: PMC10449643 DOI: 10.3389/fnagi.2023.1179086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Background Intracytoplasmic inclusions comprised of aggregated alpha-synuclein (αsyn) represent a key histopathological feature of neurological disorders collectively termed "synucleinopathies," which includes Parkinson's disease (PD). Mutations and multiplications in the SNCA gene encoding αsyn cause familial forms of PD and a large body of evidence indicate a correlation between αsyn accumulation and disease. Decreasing αsyn expression is recognized as a valid target for PD therapeutics, with down-regulation of SNCA expression potentially attenuating downstream cascades of pathologic events. Here, we evaluated if Honokiol (HKL), a polyphenolic compound derived from magnolia tree bark with demonstrated neuroprotective properties, can modulate αsyn levels in multiple experimental models. Methods Human neuroglioma cells stably overexpressing αsyn, mouse primary neurons, and human iPSC-derived neurons were exposed to HKL and αsyn protein and SNCA messenger RNA levels were assessed. The effect of HKL on rotenone-induced overexpression of αsyn levels was further assessed and transcriptional profiling of mouse cortical neurons treated with HKL was performed to identify potential targets of HKL. Results We demonstrate that HKL can successfully reduce αsyn protein levels and SNCA expression in multiple in vitro models of PD with our data supporting a mechanism whereby HKL acts by post-transcriptional modulation of SNCA rather than modulating αsyn protein degradation. Transcriptional profiling of mouse cortical neurons treated with HKL identifies several differentially expressed genes (DEG) as potential targets to modulate SNCA expression. Conclusion This study supports a HKL-mediated downregulation of SNCA as a viable strategy to modify disease progression in PD and other synucleinopathies. HKL has potential as a powerful tool for investigating SNCA gene modulation and its downstream effects.
Collapse
Affiliation(s)
- Sara J. Fagen
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Jeremy D. Burgess
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Melina J. Lim
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Danilyn Amerna
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Zeynep B. Kaya
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Ayman H. Faroqi
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Priyanka Perisetla
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Natasha N. DeMeo
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Iva Stojkovska
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Drew J. Quiriconi
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Joseph R. Mazzulli
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Marion Delenclos
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
| | - Suelen L. Boschen
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, United States
| | - Pamela J. McLean
- Department of Neuroscience, Mayo Clinic, Jackson ville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Rochester, MN, United States
| |
Collapse
|
12
|
Magnolol improves Alzheimer's disease-like pathologies and cognitive decline by promoting autophagy through activation of the AMPK/mTOR/ULK1 pathway. Biomed Pharmacother 2023; 161:114473. [PMID: 36889111 DOI: 10.1016/j.biopha.2023.114473] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. Amyloid-β (Aβ) plaque deposition and apoptosis are main pathological features of AD. Autophagy plays an important role in clearing abnormal protein accumulation and inhibiting apoptosis; however, autophagy defects often occur from the early stages of AD. The serine/threonine AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR)/unc-51-like kinase 1/2 (ULK1/2) pathway serves as an energy sensor and is involved in autophagy activation. Furthermore, magnolol is an autophagy regulator, and has potential for AD therapy. We propose that magnolol can ameliorate AD pathologies and inhibit apoptosis by regulating autophagy through the AMPK/mTOR/ULK1 pathway. We examined cognitive function and AD-related pathologies in AD transgenic mice and the protective mechanism of magnolol by western blotting, flow cytometry, and a tandem mRFP-GFP-LC3 adenovirus assay in Aβ oligomer (AβO)-induced N2a and BV2 cell models. In our study, magnolol decreased amyloid pathology and ameliorated cognitive impairment in APP/PS1 mice. Moreover, magnolol inhibited apoptosis by downregulating cleaved-caspase-9 and Bax and upregulating Bcl-2 in APP/PS1 mice and AβO-induced cell models. Magnolol promoted autophagy by degrading p62/SQSTM1, and upregulating LC3II and Beclin-1 expression. Magnolol activated the AMPK/mTOR/ULK1 pathway by increasing phosphorylation of AMPK and ULK1 and decreasing mTOR phosphorylation in in vivo and in vitro AD models. AMPK inhibitor weakened the effects of magnolol in promoting autophagy and inhibiting apoptosis, and ULK1 knockdown weakened the effect of magnolol on AβO-induced apoptosis. These results indicate that magnolol inhibits apoptosis and improves AD-related pathologies by promoting autophagy through activation of the AMPK/mTOR/ULK1 pathway.
Collapse
|
13
|
Li Q, Wang R, Zhang Z, Wang H, Lu X, Zhang J, Kong APS, Tian XY, Chan HF, Chung ACK, Cheng JCY, Jiang Q, Lee WYW. Sirt3 mediates the benefits of exercise on bone in aged mice. Cell Death Differ 2023; 30:152-167. [PMID: 36153410 PMCID: PMC9883264 DOI: 10.1038/s41418-022-01053-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 08/16/2022] [Accepted: 08/22/2022] [Indexed: 02/01/2023] Open
Abstract
Exercise in later life is important for bone health and delays the progression of osteoporotic bone loss. Osteocytes are the major bone cells responsible for transforming mechanical stimuli into cellular signals through their highly specialized lacunocanalicular networks (LCN). Osteocyte activity and LCN degenerate with aging, thus might impair the effectiveness of exercise on bone health; however, the underlying mechanism and clinical implications remain elusive. Herein, we showed that deletion of Sirt3 in osteocytes could impair the formation of osteocyte dendritic processes and inhibit bone gain in response to exercise in vivo. Mechanistic studies revealed that Sirt3 regulates E11/gp38 through the protein kinase A (PKA)/cAMP response element-binding protein (CREB) signaling pathway. Additionally, the Sirt3 activator honokiol enhanced the sensitivity of osteocytes to fluid shear stress in vitro, and intraperitoneal injection of honokiol reduced bone loss in aged mice in a dose-dependent manner. Collectively, Sirt3 in osteocytes regulates bone mass and mechanical responses through the regulation of E11/gp38. Therefore, targeting Sirt3 could be a novel therapeutic strategy to prevent age-related bone loss and augment the benefits of exercise on the senescent skeleton.
Collapse
Affiliation(s)
- Qiangqiang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Rongliang Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhe Zhang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
| | - Haixing Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaomin Lu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Jiajun Zhang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
| | - Alice Pik-Shan Kong
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hon-Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Arthur Chi-Kong Chung
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jack Chun-Yiu Cheng
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| | - Wayne Yuk-Wai Lee
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
14
|
Dai X, Xie L, Liu K, Liang Y, Cao Y, Lu J, Wang X, Zhang X, Li X. The Neuropharmacological Effects of Magnolol and Honokiol: A Review of Signal Pathways and Molecular Mechanisms. Curr Mol Pharmacol 2023; 16:161-177. [PMID: 35196977 DOI: 10.2174/1874467215666220223141101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/07/2022] [Accepted: 01/17/2022] [Indexed: 11/22/2022]
Abstract
Magnolol and honokiol are natural lignans with good physiological effects. As the main active substances derived from Magnolia officinalis, their pharmacological activities have attracted extensive attention. It is reported that both of them can cross the blood-brain barrier (BBB) and exert neuroprotective effects through a variety of mechanisms. This suggests that these two ingredients can be used as effective therapeutic compounds to treat a wide range of neurological diseases. This article provides a review of the mechanisms involved in the therapeutic effects of magnolol and honokiol in combating diseases, such as cerebral ischemia, neuroinflammation, Alzheimer's disease, and brain tumors, as well as psychiatric disorders, such as anxiety and depression. Although magnolol and honokiol have the pharmacological effects described above, their clinical potential remains untapped. More research is needed to improve the bioavailability of magnolol and honokiol and perform experiments to examine the therapeutic potential of magnolol and honokiol.
Collapse
Affiliation(s)
- Xiaolin Dai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Long Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Kai Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Youdan Liang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Yi Cao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Jing Lu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xian Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xumin Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| |
Collapse
|
15
|
Magnolia officinalis Bark Extract Prevents Enterocyte Death in a Colitis Mouse Model by Inhibiting ROS-Mediated Necroptosis. Antioxidants (Basel) 2022; 11:antiox11122435. [PMID: 36552643 PMCID: PMC9774795 DOI: 10.3390/antiox11122435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Necroptosis is a form of programmed cell death with features of necrosis and apoptosis that occurs in the intestinal epithelium of patients with inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease. In addition, necroptosis has also been observed in enterocytes in animal models of dextran sulfate sodium (DSS)-induced colitis. Thus, the discovery of natural products for regulating necroptosis may represent an important therapeutic strategy for improving IBD. We found that Magnolia officinalis bark extract (MBE) prevented weight loss and suppressed the activation of the proinflammatory cytokine IL6 in DSS-induced colitis. Furthermore, MBE restored the length of the damaged colon and decreased the expression of necroptosis markers in mice with DSS-induced colitis. In vitro, necroptosis-induced reactive oxygen species (ROS) production was reduced by MBE, and the expression of COX2, a target protein of ROS, was simultaneously suppressed. Both magnolol and honokiol, the two major bioactive compounds in MBE, inhibited necroptosis in human primary intestinal epithelial cells and colorectal adenocarcinoma cells. Our findings highlight the effectiveness of MBE in modulating enterocyte necroptosis and suggest that MBE may be developed as a natural, disease-targeting drug for the treatment of colitis.
Collapse
|
16
|
Calderon-Rivera A, Loya-Lopez S, Gomez K, Khanna R. Plant and fungi derived analgesic natural products targeting voltage-gated sodium and calcium channels. Channels (Austin) 2022; 16:198-215. [PMID: 36017978 PMCID: PMC9423853 DOI: 10.1080/19336950.2022.2103234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Voltage-gated sodium and calcium channels (VGSCs and VGCCs) play an important role in the modulation of physiologically relevant processes in excitable cells that range from action potential generation to neurotransmission. Once their expression and/or function is altered in disease, specific pharmacological approaches become necessary to mitigate the negative consequences of such dysregulation. Several classes of small molecules have been developed with demonstrated effectiveness on VGSCs and VGCCs; however, off-target effects have also been described, limiting their use and spurring efforts to find more specific and safer molecules to target these channels. There are a great number of plants and herbal preparations that have been empirically used for the treatment of diseases in which VGSCs and VGCCs are involved. Some of these natural products have progressed to clinical trials, while others are under investigation for their action mechanisms on signaling pathways, including channels. In this review, we synthesize information from ~30 compounds derived from natural sources like plants and fungi and delineate their effects on VGSCs and VGCCs in human disease, particularly pain. [Figure: see text].
Collapse
Affiliation(s)
- Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA,NYU Pain Research Center, New York University, New York, NY, USA
| | - Santiago Loya-Lopez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA,NYU Pain Research Center, New York University, New York, NY, USA
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA,NYU Pain Research Center, New York University, New York, NY, USA
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA,NYU Pain Research Center, New York University, New York, NY, USA,CONTACT Rajesh Khanna
| |
Collapse
|
17
|
Jarne-Ferrer J, Griñán-Ferré C, Bellver-Sanchis A, Vázquez S, Muñoz-Torrero D, Pallàs M. A Combined Chronic Low-Dose Soluble Epoxide Hydrolase and Acetylcholinesterase Pharmacological Inhibition Promotes Memory Reinstatement in Alzheimer’s Disease Mice Models. Pharmaceuticals (Basel) 2022; 15:ph15080908. [PMID: 35893732 PMCID: PMC9394299 DOI: 10.3390/ph15080908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/04/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurological disorder with multifactorial and heterogeneous causes. AD involves several etiopathogenic mechanisms such as aberrant protein accumulation, neurotransmitter deficits, synaptic dysfunction and neuroinflammation, which lead to cognitive decline. Unfortunately, the currently available anti-AD drugs only alleviate the symptoms temporarily and provide a limited therapeutic effect. Thus, new therapeutic strategies, including multitarget approaches, are urgently needed. It has been demonstrated that a co-treatment of acetylcholinesterase (AChE) inhibitor with other neuroprotective agents has beneficial effects on cognition. Here, we have assessed the neuroprotective effects of chronic dual treatment with a soluble epoxide hydrolase (sEH) inhibitor (TPPU) and an AChE inhibitor (6-chlorotacrine or rivastigmine) in in vivo studies. Interestingly, we have found beneficial effects after chronic low-dose co-treatment with TPPU and 6-chlorotacrine in the senescence-accelerated mouse prone 8 (SAMP8) mouse model as well as with TPPU and rivastigmine co-treatment in the 5XFAD mouse model, in comparison with the corresponding monotherapy treatments. In the SAMP8 model, no substantial improvements in synaptic plasticity markers were found, but the co-treatment of TPPU and 6-chlorotacrine led to a significantly reduced gene expression of neuroinflammatory markers, such as interleukin 6 (Il-6), triggering receptor expressed on myeloid cell 2 (Trem2) and glial fibrillary acidic protein (Gfap). In 5XFAD mice, chronic low-dose co-treatment of TPPU and rivastigmine led to enhanced protein levels of synaptic plasticity markers, such as the phospho-cAMP response element-binding protein (p-CREB) ratio, brain-derived neurotrophic factor (BDNF) and postsynaptic density protein 95 (PSD95), and also to a reduction in neuroinflammatory gene expression. Collectively, these results support the neuroprotectant role of chronic low-dose co-treatment strategy with sEH and AChE inhibitors in AD mouse models, opening new avenues for effective AD treatment.
Collapse
Affiliation(s)
- Júlia Jarne-Ferrer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, Avenida Joan XXIII, 27-31, E-08028 Barcelona, Spain; (J.J.-F.); (C.G.-F.); (A.B.-S.)
| | - Christian Griñán-Ferré
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, Avenida Joan XXIII, 27-31, E-08028 Barcelona, Spain; (J.J.-F.); (C.G.-F.); (A.B.-S.)
| | - Aina Bellver-Sanchis
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, Avenida Joan XXIII, 27-31, E-08028 Barcelona, Spain; (J.J.-F.); (C.G.-F.); (A.B.-S.)
| | - Santiago Vázquez
- CSIC Associated Unit, Laboratory of Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Avenida Joan XXIII, 27-31, E-08028 Barcelona, Spain; (S.V.); (D.M.-T.)
| | - Diego Muñoz-Torrero
- CSIC Associated Unit, Laboratory of Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Avenida Joan XXIII, 27-31, E-08028 Barcelona, Spain; (S.V.); (D.M.-T.)
| | - Mercè Pallàs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, Avenida Joan XXIII, 27-31, E-08028 Barcelona, Spain; (J.J.-F.); (C.G.-F.); (A.B.-S.)
- Correspondence:
| |
Collapse
|
18
|
Zhu S, Liu F, Zhang R, Xiong Z, Zhang Q, Hao L, Chen S. Neuroprotective Potency of Neolignans in Magnolia officinalis Cortex Against Brain Disorders. Front Pharmacol 2022; 13:857449. [PMID: 35784755 PMCID: PMC9244706 DOI: 10.3389/fphar.2022.857449] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/20/2022] [Indexed: 12/03/2022] Open
Abstract
In recent years, neurological diseases including Alzheimer’s disease, Parkinson’s disease and stroke are one of the main causes of death in the world. At the same time, the incidence of psychiatric disorders including depression and anxiety has been increasing. Accumulating elderly and stressed people suffer from these brain disorders, which is undoubtedly a huge burden on the modern aging society. Neolignans, the main active ingredients in Magnolia officinalis cortex, were reported to have neuroprotective effects. In addition, the key bioactive ingredients of neolignans, magnolol (1) and honokiol (2), were proved to prevent and treat neurological diseases and psychiatric disorders by protecting nerve cells and brain microvascular endothelial cells (BMECs). Furthermore, neolignans played a role in protecting nerve cells via regulation of neuronal function, suppression of neurotoxicity, etc. This review summarizes the neuroprotective effect, primary mechanisms of the leading neolignans and provides new prospects for the treatment of brain disorders in the future.
Collapse
Affiliation(s)
- Shun Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Fang Liu, ; Shiyin Chen,
| | - Ruiyuan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zongxiang Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Hao
- Huarun Sanjiu (ya’an) Pharmaceutical Group Co., LTD., Ya’an, China
| | - Shiyin Chen
- Department of Orthopedics of Traditional Chinese Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Fang Liu, ; Shiyin Chen,
| |
Collapse
|
19
|
Bibi T, Khan A, Khan AU, Shal B, Ali H, Seo EK, Khan S. Magnolol prevented brain injury through the modulation of Nrf2-dependent oxidative stress and apoptosis in PLP-induced mouse model of multiple sclerosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:717-733. [PMID: 35348816 DOI: 10.1007/s00210-022-02230-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/11/2022] [Indexed: 01/07/2023]
Abstract
Multiple sclerosis (MS) is an immune-mediated chronic inflammatory demyelinating disease of the central nervous system (CNS). The aim of the current study was to investigate the effects of magnolol in an experimental autoimmune encephalomyelitis (EAE) model of MS in female mice. Magnolol (0.1, 1, and 10 mg/kg) was administered once daily for 21 days after immunization of mice. Magnolol post-immunization treatment significantly reversed clinical scoring, EAE-associated pain parameters, and motor dysfunction in a dose-dependent manner. Magnolol treatment significantly inhibited oxidative stress by reducing malondialdehyde (MDA), nitric oxide (NO) production, and myeloperoxidase (MPO) activity while enhancing the level of antioxidants such as reduced glutathione (GSH), glutathione-S-transferase (GST), catalase, and superoxide dismutase (SOD) in the brain and spinal cord. It reduced cytokine levels in the brain and spinal cord. It suppressed CD8+ T cells frequency in the spleen tissue. Magnolol remarkably reversed the EAE-associated histopathology of the brain and spinal cord tissue. Magnolol significantly intensifies the antioxidant defense system by enhancing the expression level of nuclear factor erythroid 2-related factor (Nrf2) while decreasing the expression of inducible nitric oxide synthase (iNOS) and cleaved-caspase-3 in the brain. Molecular docking results showed that magnolol possesses a better binding affinity for Nrf2, iNOS, and caspase-3 proteins. Taken together, the present study demonstrated that magnolol has significant neuroprotective properties in EAE via inhibition of oxidative stress.
Collapse
Affiliation(s)
- Tehmina Bibi
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Adnan Khan
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ashraf Ullah Khan
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Faculty of Pharmaceutical Sciences, Abasyn University, Peshawar, Pakistan
| | - Bushra Shal
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Faculty of Health Sciences, IQRA University, Islamabad Campus, (Chak Shahzad), Islamabad, Pakistan
| | - Hussain Ali
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Eun Kyoung Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea.
| | - Salman Khan
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
20
|
Qu C, Li QP, Su ZR, Ip SP, Yuan QJ, Xie YL, Xu QQ, Yang W, Huang YF, Xian YF, Lin ZX. Nano-Honokiol ameliorates the cognitive deficits in TgCRND8 mice of Alzheimer's disease via inhibiting neuropathology and modulating gut microbiota. J Adv Res 2022; 35:231-243. [PMID: 35024199 PMCID: PMC8721355 DOI: 10.1016/j.jare.2021.03.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/08/2021] [Accepted: 03/28/2021] [Indexed: 01/05/2023] Open
Abstract
Introduction Honokiol (HO) exerts neuroprotective effects in several animal models of Alzheimer's disease (AD), but the poor dissolution hampers its bioavailability and therapeutic efficacy. Objectives A novel honokiol nanoscale drug delivery system (Nano-HO) with smaller size and excellent stability was developed in this study to improve the solubility and bioavailability of HO. The anti-AD effects of Nano-HO was determined. Methods Male TgCRND8 mice were daily orally administered Nano-HO or HO at the same dosage (20 mg/kg) for 17 consecutive weeks, followed by assessment of the spatial learning and memory functions using the Morris Water Maze test (MWMT). Results Our pharmacokinetic study indicated that the oral bioavailability was greatly improved by Nano-HO. In addition, Nano-HO significantly improved cognitive deficits and inhibited neuroinflammation via suppressing the levels of TNF-α, IL-6 and IL-1β in the brain, preventing the activation of microglia (IBA-1) and astrocyte (GFAP), and reducing β-amyloid (Aβ) deposition in the cortex and hippocampus of TgCRND8 mice. Moreover, Nano-HO was more effective than HO in modulating amyloid precursor protein (APP) processing via suppressing β-secretase, as well as enhancing Aβ-degrading enzymes like neprilysin (NEP). Furthermore, Nano-HO more markedly inhibited tau hyperphosphorylation via decreasing the ratio of p-Tau (Thr 205)/tau and regulating tau-related apoptosis proteins (caspase-3 and Bcl-2). In addition, Nano-HO more markedly attenuated the ratios of p-JNK/JNK and p-35/CDK5, while enhancing the ratio of p-GSK-3β (Ser9)/GSK-3β. Finally, Nano-HO prevented the gut microflora dysbiosis in TgCRND8 mice in a more potent manner than free HO. Conclusion Nano-HO was more potent than free HO in improving cognitive impairments in TgCRND8 mice via inhibiting Aβ deposition, tau hyperphosphorylation and neuroinflammation through suppressing the activation of JNK/CDK5/GSK-3β signaling pathway. Nano-HO also more potently modulated the gut microbiota community to protect its stability than free HO. These results suggest that Nano-HO has good potential for further development into therapeutic agent for AD treatment.
Collapse
Key Words
- AD, Alzheimer’s disease
- APH-1, anterior pharynx-defective-1
- APP, amyloid precursor protein
- Aβ, β-amyloid
- BACE-1, β-site APP cleaving enzyme-1
- Bcl-2, B cell lymphoma-2
- CDK5, cyclin-dependent kinase 5
- CMC-Na, sodium carboxymethylcellulose
- Cognitive deficits
- GSK-3β, glycogen synthase kinase 3β
- Gut microbiota
- HO, Honokiol
- HPLC, high performance liquid chromatography
- Honokiol nanoscale drug delivery system
- IDE, insulin degrading enzyme
- IL-1β, interleukin 1β
- IL-6, interleukin 6
- JNK, c-Jun N-terminal kinase
- MCT, Medium-chain triglycerides
- MWMT, Morris Water Maze test
- NEP, neprilysin
- NFTs, neurofibrillary tangles
- Nano-HO, honokiol nanoscale drug delivery system
- Neuroinflammation
- PBS, phosphate-buffered saline
- PDI, poly-dispersity index
- PS-1, presenilin-1
- ROS, reactive oxygen species
- TEM, transmission electron microscope
- TNF-α, tumor necrosis factor
- Tau protein hyperphosphorylation
- TgCRND8 mice
- WT, wild type
- ZP, zeta potential
Collapse
Affiliation(s)
- Chang Qu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Qiao-Ping Li
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zi-Ren Su
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Siu-Po Ip
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.,Brain Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Qiu-Ju Yuan
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.,Brain Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - You-Liang Xie
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qing-Qing Xu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Wen Yang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Yan-Feng Huang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.,Brain Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.,Brain Research Centre, School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.,Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| |
Collapse
|
21
|
Niu L, Hou Y, Jiang M, Bai G. The rich pharmacological activities of Magnolia officinalis and secondary effects based on significant intestinal contributions. JOURNAL OF ETHNOPHARMACOLOGY 2021; 281:114524. [PMID: 34400262 DOI: 10.1016/j.jep.2021.114524] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/01/2021] [Accepted: 08/13/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Magnolia officinalis Cortex (M. officinalis) is a traditional herbal drug widely used in Asian countries. Depending on its multiple biological activities, M. officinalis is used to regulate gastrointestinal (GI) motility, relieve cough and asthma, prevent cardiovascular and cerebrovascular diseases, and treat depression and anxiety. AIM OF THE REVIEW We aimed to review the abundant form of pharmacodynamics activity and potential mechanisms of action of M. officinalis and the characteristics of the internal processes of the main components. The potential mechanisms of local and distance actions of M. officinalis based on GI tract was provided, and it was used to reveal the interconnections between traditional use, phytochemistry, and pharmacology. MATERIALS AND METHODS Published literatures about M. officinalis and its main components were collected from several scientific databases, including PubMed, Elsevier, ScienceDirect, Google Scholar and Web of Science etc. RESULTS: M. officinalis was shown multiple effects including effects on digestive system, respiratory system, central system, which is consistent with traditional applications, as well as some other activities such as cardiovascular system, anticancer, anti-inflammatory and antioxidant effects and so on. The mechanisms of these activities are abundant. Its chief ingredients such as magnolol and honokiol can be metabolized into active metabolites in vivo, which can increase water solubility and bioavailability and exert pharmacological activity in the whole body. In the GI tract, M. officinalis and its main ingredient can regulate GI hormones and substance metabolism, protect the intestinal barrier and affect the gut microbiota (GM). These actions are effective to improve local discomfort and some distal symptoms such as depression, asthma, or metabolic disorders. CONCLUSIONS Although M. officinalis has rich pharmacological effects, the GI tract makes great contributions to it. The GI tract is not only an important place for absorption and metabolism but also a key site to help M. officinalis exert local and distal efficacy. Pharmacodynamical studies on the efficacies of distal tissues based on the contributions of the GI tract hold great potential for understanding the benefits of M. officinalis and providing new ideas for the treatment of important diseases.
Collapse
Affiliation(s)
- Lin Niu
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Min Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China.
| |
Collapse
|
22
|
Zhang X, Wang J, Sui A, Zhang N, Lv Q, Liu Z. Antinociceptive Effect of Magnolol in a Neuropathic Pain Model of Mouse. J Pain Res 2021; 14:2083-2093. [PMID: 34267552 PMCID: PMC8275150 DOI: 10.2147/jpr.s317204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/23/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Neuropathic pain remains a clinical challenge with limited effective treatments. Previous studies have found that magnolol (Mag), an ingredient existing in some herbs, showed neuroprotective effect. However, it remains unclear whether Mag can alleviate neuropathic pain. METHODS Chronic constriction injury (CCI) is used as the neuropathic pain model. Mice were randomly divided into 5 groups: Sham, CCI, CCI + 5, 10, 30 mg/kg Mag groups. Thermal and mechanical paw withdrawal threshold were performed at baseline and on the 3rd, 5th, 7th, 14th days post-surgery. Lumbar spinal cord and blood samples were collected on the 14th day. Blood lipid profile, kidney and liver functions, as well as the activation of microglia were evaluated, along with the related signal pathway examined using multiple methods including immunohistochemistry, RT-PCR and Western blot. RESULTS Mag alleviated thermal and mechanical hypersensitivity in CCI mice. CCI activated microglia and upregulated the expression of P2Y12, while Mag inhibited microglial activation, and downregulated the expression of P2Y12. Mag also blocked the activation of p38 mitogen-activated protein kinase (MAPK) and other pain-related cytokines such as IL-6, TNF-α and IL-1β. CONCLUSION The findings indicate that Mag has antinociceptive effect on neuropathic pain, probably mediated through P2Y12 receptors and p38 MAPK mediated pathways. With its relatively safe profile, Mag may be a potential therapeutic agent for neuropathic pain.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Juntao Wang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Aihua Sui
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Nannan Zhang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Qiulan Lv
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| | - Zhenfang Liu
- Department of Emergency, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China
| |
Collapse
|
23
|
Chen C, Zhang QW, Ye Y, Lin LG. Honokiol: A naturally occurring lignan with pleiotropic bioactivities. Chin J Nat Med 2021; 19:481-490. [PMID: 34247771 DOI: 10.1016/s1875-5364(21)60047-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Indexed: 12/16/2022]
Abstract
Honokiol is the dominant biphenolic compound isolated from the Magnolia tree, and has long been considered as the active constituent of the traditional Chinese herb, 'Houpo', which is widely used to treat symptoms due to 'stagnation of qi'. Pharmacological studies have shown that honokiol possesses a wide range of bioactivities without obvious toxicity. Honokiol protects the liver, kidneys, nervous system, and cardiovascular system through reducing oxidative stress and relieving inflammation. Moreover, honokiol shows anti-diabetic property through enhancing insulin sensitivity, and anti-obese property through promoting browning of adipocytes. In vivo and in vitro studies indicated that honokiol functions as an anti-cancer agent through multiple mechanisms: inhibiting angiogenesis, promoting cell apoptosis, and regulating cell cycle. A variety of therapeutic effects of honokiol may be associated with its physiochemical properties, which make honokiol readily cross the blood brain barrier and the blood-cerebrospinal fluid barrier, with high bioavailability. In the future, more clinical researches on honokiol are needed to fully authenticate its therapeutic values.
Collapse
Affiliation(s)
- Cheng Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Qing-Wen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yang Ye
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Li-Gen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
24
|
Zweig JA, Brandes MS, Brumbach BH, Caruso M, Wright KM, Quinn JF, Soumyanath A, Gray NE. Loss of NRF2 accelerates cognitive decline, exacerbates mitochondrial dysfunction, and is required for the cognitive enhancing effects of Centella asiatica during aging. Neurobiol Aging 2021; 100:48-58. [PMID: 33486357 PMCID: PMC7920997 DOI: 10.1016/j.neurobiolaging.2020.11.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
Abstract
The water extract of Centella asiatica (CAW) improves cognitive and mitochondrial function and activates the nuclear factor erythroid 2-related factor 2 (NRF2) regulated antioxidant response pathway in aged mice. Here we investigate whether NRF2 activation is required for the cognitive and mitochondrial effects of prolonged CAW exposure during aging. Five-month-old NRF2 knockout (NRF2KO) and wild-type mice were treated with CAW for 1, 7, or 13 months. Each cohort underwent cognitive testing and hippocampal mitochondrial analyses. Age-related cognitive decline was accelerated in NRF2KO mice and while CAW treatment improved cognitive performance in wild-type mice, it had no effect on NRF2KO animals. Hippocampal mitochondrial function also declined further with age in NRF2KO mice and greater hippocampal mitochondrial dysfunction was associated with poorer cognitive performance in both genotypes. Long-term CAW treatment did not affect mitochondrial endpoints in animals of either genotype. These data indicate that loss of NRF2 results in accelerated age-related cognitive decline and worsened mitochondrial deficits. NRF2 also appears to be required for the cognitive enhancing effects of CAW during aging.
Collapse
Affiliation(s)
- Jonathan A Zweig
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Mikah S Brandes
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Barbara H Brumbach
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, USA
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Kirsten M Wright
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA; Department of Neurology and Parkinson's Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, OR, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Nora E Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
25
|
Soheili M, Karimian M, Hamidi G, Salami M. Alzheimer's disease treatment: The share of herbal medicines. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:123-135. [PMID: 33953850 PMCID: PMC8061323 DOI: 10.22038/ijbms.2020.50536.11512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/07/2020] [Indexed: 11/25/2022]
Abstract
One of the most frequent forms of dementia in neurological disorders is Alzheimer's disease (AD). It is a chronic neurodegenerative disease characterized by impaired learning and memory. Pathological symptoms as extracellular amyloid-beta (Aβ) plaques and intracellular accumulation of neurofibrillary tangles occur in AD. Due to the aging of the population and increased prevalence of AD, discovery of new therapeutic agents with the highest effectiveness and fewer side effect seems to be necessary. Numerous synthetic medicines such as tacrine, donepezil, galantamine, rivastigmine, memantine, glutathione, ascorbic acid, ubiquinone, ibuprofen, and ladostigil are routinely used for reduction of the symptoms and prevention of disease progression. Nowadays, herbal medicines have attracted popular attention for numerous beneficial effects with little side effects. Lavandula angustifolia, Ginkgo biloba, Melissa officinalis, Crocus sativus, Ginseng, Salvia miltiorrhiza, and Magnolia officinalis have been widely used for relief of symptoms of some neurological disorders. This paper reviews the therapeutic effects of phytomedicines with prominent effects against various factors implicated in the emergence and progression of AD.
Collapse
Affiliation(s)
- Masoud Soheili
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Gholamali Hamidi
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahmoud Salami
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
26
|
Govindarajulu M, Ramesh S, Neel L, Fabbrini M, Buabeid M, Fujihashi A, Dwyer D, Lynd T, Shah K, Mohanakumar KP, Smith F, Moore T, Dhanasekaran M. Nutraceutical based SIRT3 activators as therapeutic targets in Alzheimer's disease. Neurochem Int 2021; 144:104958. [PMID: 33444675 DOI: 10.1016/j.neuint.2021.104958] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 12/21/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, and its incidence is increasing worldwide with increased lifespan. Currently, there is no effective treatment to cure or prevent the progression of AD, which indicates the need to develop novel therapeutic targets and agents. Sirtuins, especially SIRT3, a mitochondrial deacetylase, are NAD-dependent histone deacetylases involved in aging and longevity. Accumulating evidence indicates that SIRT3 dysfunction is strongly associated with pathologies of AD, hence, therapeutic modulation of SIRT3 activity may be a novel application to ameliorate the pathologies of AD. Natural products commonly used in traditional medicine have wide utility and appear to have therapeutic benefits for the treatment of neurodegenerative diseases such as AD. The present review summarizes the currently available natural SIRT3 activators and their potentially neuroprotective molecular mechanisms of action that make them a promising agent in the treatment and management of neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Manoj Govindarajulu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Sindhu Ramesh
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Logan Neel
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Mary Fabbrini
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Manal Buabeid
- Clinical Pharmacy Department, College of Pharmacy and Health Sciences, Ajman University, United Arab Emirates
| | - Ayaka Fujihashi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Darby Dwyer
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Tyler Lynd
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Karishma Shah
- Department of Ophthalmology, D.Y. Patil Medical College and Research Hospital, Mumbai, India
| | | | - Forrest Smith
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Timothy Moore
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, AL 36849, USA.
| |
Collapse
|
27
|
Fukuyama Y, Kubo M, Harada K. The search for, and chemistry and mechanism of, neurotrophic natural products. J Nat Med 2020; 74:648-671. [PMID: 32643028 PMCID: PMC7456418 DOI: 10.1007/s11418-020-01431-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 06/28/2020] [Indexed: 12/13/2022]
Abstract
Abstract Neurotrophic factors, now termed neurotrophins, which belong to a class of polypeptidyl agents, have been shown to potentially be beneficial for the treatment of neurodegenerative diseases such as Alzheimer’s disease, because endogenous neurotrophic factors (NGF, BDNF, NT3, NT4) have been recognized to play critical roles in the promotion of neurogenesis, differentiation, and neuroprotection throughout the development of the central nervous system. However, high-molecular weight proteins are unable to cross the blood–brain barrier and are easily decomposed by peptidase under physiological conditions. To address this issue, small molecules that can mimic the functions of neurotrophic factors would be promising alternatives for the treatment of neurodegenerative disease. We have continued to search for natural products having typical neurotrophic properties, which can cause neurogenesis, enhance neurite outgrowth, and protect neuronal death using three cellular systems (PC12, rat cortical neurons, and MEB5 cells). In this review, we summarize the neurotrophic activities and synthesis of dimeric isocuparane-type sesquiterpenes from the liverwort, Mastigophora diclados, the mechanism of neurotrophic neolignans, magnolol, honokiol and their sesquiterpene derivatives, and introduce unique neurotrophin-mimic natural products, including seco-prezizaane-type sesquiterpenes from the Illicium species, vibsane-type diterpenes from Viburnum awabuki, and miscellaneous natural products with neurotrophic effects discovered by us. Graphic abstract ![]()
Collapse
Affiliation(s)
- Yoshiyasu Fukuyama
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan.
| | - Miwa Kubo
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan
| | - Kenichi Harada
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan
| |
Collapse
|
28
|
Magnolol Ameliorates Behavioral Impairments and Neuropathology in a Transgenic Mouse Model of Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5920476. [PMID: 32714487 PMCID: PMC7354664 DOI: 10.1155/2020/5920476] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/13/2020] [Indexed: 12/02/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease characterized by progressive memory loss. Magnolol (MN), the main active ingredient of Magnolia officinalis, possesses anti-AD effects in several experimental models of AD. In this study, we aimed to explore whether MN could ameliorate the cognitive deficits in TgCRND8 transgenic mice and to elucidate its molecular mechanisms. Male TgCRND8 mice were orally administered with MN (20 and 40 mg/kg) daily for 4 consecutive months, followed by assessing the spatial learning and memory functions using the open-field, radial arm maze, and novel object recognition tests. The results demonstrated that MN (20 and 40 mg/kg) could markedly ameliorate the cognitive deficits in TgCRND8 mice. In addition, MN significantly increased the expression of postsynaptic density protein 93 (PSD93), PSD-95, synapsin-1, synaptotagmin-1, synaptophysin (SYN), and interleukin-10 (IL-10), while markedly reduced the protein levels of tumor necrosis factor alpha (TNF-α), IL-6, IL-1β, Aβ40, and Aβ42, and modulated the amyloid precursor protein (APP) processing and phosphorylation. Immunofluorescence showed that MN significantly suppressed the activation of microglia (Iba-1) and astrocytes (GFAP) in the hippocampus and cerebral cortex of TgCRND8 mice. Mechanistic studies revealed that MN could significantly increase the ratios of p-GSK-3β (Ser9)/GSK-3β, p-Akt (Ser473)/Akt, and p-NF-κB p65/NF-κB p65. These findings indicate that MN exerted cognitive deficits improving effects via suppressing neuroinflammation, amyloid pathology, and synaptic dysfunction through regulating the PI3K/Akt/GSK-3β and NF-κB pathways, suggesting that MN is a promising naturally occurring polyphenol worthy of further developing into a therapeutic agent for AD treatment.
Collapse
|
29
|
Kim A, Lee SY, Seo CS, Chung SK. Ethanol extract of Magnoliae cortex (EEMC) limits teratoma formation of pluripotent stem cells by selective elimination of undifferentiated cells through the p53-dependent mitochondrial apoptotic pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 69:153198. [PMID: 32151917 DOI: 10.1016/j.phymed.2020.153198] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 01/16/2020] [Accepted: 02/25/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs) are regarded as the best potential cell source for cell-based regenerative medicine. To develop a safe and efficient iPSC-based cell therapy, it is very important to avoid possible teratoma formation, which can arise from undifferentiated iPSCs (USCs) remaining among differentiated cell products. Dried bark of Magnolia officinalis (Magnolia cortex, MC) has long been used in traditional medicine to treat gastrointestinal ailments and allergic diseases, and has shown have various pharmacological activities, including anti-bacterial, anti-inflammatory, and anti-cancer effects. However, its effects on iPSCs have not yet been examined. PURPOSE In this study, we investigated the selective cytotoxic effects of ethanol extract of MC (EEMC) on undifferentiated iPSCs and elucidated the underlying apoptotic mechanisms in detail. We also investigated the inhibitory effects of EEMC on teratoma formation via in ovo experiments. RESULTS We found that EEMC greatly reduced cell growth and induced apoptotic cell death in USCs, but not in differentiated or normal cells. EEMC caused G2/M cell cycle arrest, mitochondrial damage, and caspase activation of USCs, accompanied by p53 accumulation. In p53KO human iPSCs, EEMC had no cytotoxicity, reinforcing that EEMC-mediated apoptosis of USCs is p53-dependent. EEMC did not cause DNA damage in iPSC-derived differentiated cells. In ovo teratoma formation assay revealed that EEMC treatment before injection efficiently eliminated USCs and prevented teratoma formation. CONCLUSIONS These results collectively indicate that EEMC has potent anti-teratoma activity, and therefore can be used for the development of safe iPSC-based therapy.
Collapse
Affiliation(s)
- Aeyung Kim
- Division of Clinical Medicine, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea.
| | - Seo-Young Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Chang-Seob Seo
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Sun-Ku Chung
- Division of Clinical Medicine, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea.
| |
Collapse
|
30
|
Zweig JA, Caruso M, Brandes MS, Gray NE. Loss of NRF2 leads to impaired mitochondrial function, decreased synaptic density and exacerbated age-related cognitive deficits. Exp Gerontol 2019; 131:110767. [PMID: 31843395 DOI: 10.1016/j.exger.2019.110767] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/15/2019] [Accepted: 10/29/2019] [Indexed: 12/14/2022]
Abstract
Activation of the antioxidant regulatory transcription factor NRF2 (Nuclear factor erythroid-derived 2) regulates cellular bioenergetics and improves neuronal health in aging. Yet how NRF2 participates in maintaining synaptic, mitochondrial and cognitive function has not been fully elucidated. This study investigates how loss of NRF2 affects neuronal metabolism, synaptic density and cognitive performance in aged mice. Dendritic arborization as well as synaptic and mitochondrial gene expression was evaluated in hippocampal neurons isolated from mice lacking NRF2 (NRF2KO) and from wild-type (WT) C57BL6 mice. Mitochondrial function of these neurons was evaluated using the Seahorse XF platform. Additionally learning, memory and executive function were assessed in 20 month old NRF2KO and age-matched WT mice using conditioned fear response (CFR) and odor discrimination reversal learning (ODRL) tests. Hippocampal bioenergetics was profiled using mitochondria isolated from these animals and tissue was harvested for assessment of mitochondrial and synaptic genes. NRF2KO neurons had reduced dendritic complexity and diminished synaptic gene expression. This was accompanied by impaired mitochondrial function and decreased mitochondrial gene expression. Similar mitochondrial deficits were observed in the brains of aged NRF2KO mice. These animals also had significantly impaired cognitive performance and reduced synaptic gene expression as well. These data point to a role for NRF2 in maintaining mitochondrial and cognitive function during aging and suggest that the transcription factor may be a viable target for cognitive enhancing interventions. Because mitochondrial dysfunction and cognitive impairment also occur together in many neurodegenerative conditions there may be broad therapeutic potential of NRF2 activating agents.
Collapse
Affiliation(s)
- Jonathan A Zweig
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Maya Caruso
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Mikah S Brandes
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
31
|
Guo S, Xu JJ, Wei N, Han JY, Xue R, Xu PS, Gao CY. Honokiol Attenuates the Memory Impairments, Oxidative Stress, Neuroinflammation, and GSK-3β Activation in Vascular Dementia Rats. J Alzheimers Dis 2019; 71:97-108. [PMID: 31322570 DOI: 10.3233/jad-190324] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Si Guo
- Department of Medical Laboratory, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
- Department of Medical Laboratory, Fuwai Central China Cardiovascular Hospital, Zhengzhou, People’s Republic of China
| | - Jing-Jing Xu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou, People’s Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Na Wei
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou, People’s Republic of China
- Department of Pathology, School of Basic Medicine, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Jun-Ya Han
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Po-Shi Xu
- Department of Medical Laboratory, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
- Department of Medical Laboratory, Fuwai Central China Cardiovascular Hospital, Zhengzhou, People’s Republic of China
| | - Chuan-Yu Gao
- Department of Cardiology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
- Department of Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, People’s Republic of China
| |
Collapse
|
32
|
Barua S, Kim JY, Yenari MA, Lee JE. The role of NOX inhibitors in neurodegenerative diseases. IBRO Rep 2019; 7:59-69. [PMID: 31463415 PMCID: PMC6709343 DOI: 10.1016/j.ibror.2019.07.1721] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/27/2019] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is a key player in both chronic and acute brain disease due to the higher metabolic demand of the brain. Among the producers of free radicals, NADPH-oxidase (NOX) is a major contributor to oxidative stress in neurological disorders. In the brain, the superoxide produced by NOX is mainly found in leukocytes. However, recent studies have reported that it can be found in several other cell types. NOX has been reported to regulate neuronal signaling, memory processing, and central cardiovascular homeostasis. However, overproduction of NOX can contribute to neurotoxicity, CNS degeneration, and cardiovascular disorders. Regarding the above functions, NOX has been shown to play a crucial role in chronic CNS diseases like Parkinson's disease (PD), Alzheimer's disease (AD), Huntington's disease (HD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS), and in acute CNS disorders such as stroke, spinal cord injury, traumatic brain injury (TBI), and related cerebrovascular diseases. NOX is a multi-subunit complex consisting of two membrane-associated and four cytosolic subunits. Thus, in recent years, inhibition of NOX activity has drawn a great deal of attention from researchers in the field of treating chronic and acute CNS disorders and preventing secondary complications. Mounting evidence has shown that NOX inhibition is neuroprotective and that inhibiting NOX in circulating immune cells can improve neurological disease conditions. This review summarizes recent studies on the therapeutic effects and pharmacological strategies regarding NOX inhibitors in chronic and acute brain diseases and focuses on the hurdles that should be overcome before their clinical implementation.
Collapse
Affiliation(s)
- Sumit Barua
- Department of Anatomy, College of Medicine, Yonsei University, Republic of Korea
| | - Jong Youl Kim
- Department of Anatomy, College of Medicine, Yonsei University, Republic of Korea
| | - Midori A Yenari
- Department of Neurology, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, 4150 Clement Street, MS 127, San Francisco, CA, 94121, United States
| | - Jong Eun Lee
- Department of Anatomy, College of Medicine, Yonsei University, Republic of Korea.,Brain Korea 21, PLUS Project for Medical Science, College of Medicine, Yonsei University, Republic of Korea.,Brain Research Institute, College of Medicine, Yonsei University, Republic of Korea
| |
Collapse
|
33
|
Li H, Jia J, Wang W, Hou T, Tian Y, Wu Q, Xu L, Wei Y, Wang X. Honokiol Alleviates Cognitive Deficits of Alzheimer's Disease (PS1V97L) Transgenic Mice by Activating Mitochondrial SIRT3. J Alzheimers Dis 2019; 64:291-302. [PMID: 29865070 DOI: 10.3233/jad-180126] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Accumulating evidence has demonstrated that mitochondrial dysfunction is a prominent early event in the progression of Alzheimer's disease (AD). Whether protecting mitochondrial function can reduce amyloid-β oligomer (AβO)-induced neurotoxicity in PS1V97L transgenic mice remains unknown. In this study, we examined the possible protective effects of honokiol (HKL) on mitochondrial dysfunction induced by AβOs in neurons, and cognitive function in AD PS1V97Ltransgenic mice. We determined that HKL increased mitochondrial sirtuin 3 (SIRT3) expression levels and activity, which in turn markedly improved ATP production and weakened mitochondrial reactive oxygen species production. We demonstrated that the enhanced energy metabolism and attenuated oxidative stress of HKL restores AβO-mediated mitochondrial dysfunction in vitro and in vivo. Consequently, memory deficits in the PS1V97L transgenic mice were rescued by HKL in the early stages. These results suggest that HKL has therapeutic potential for delaying the onset of AD symptoms by alleviating mitochondrial impairment and increasing hyperactivation of SIRT3 in the pathogenesis of preclinical AD.
Collapse
Affiliation(s)
- Haitao Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, P.R. China
| | - Jianping Jia
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, P.R. China.,Clinical Center for Neurodegenerative Disease and MemoryImpairment, Capital Medical University, Beijing, P.R. China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.,National Clinical Research Center forGeriatric Disorders, Beijing, P.R. China
| | - Wei Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, P.R. China.,Clinical Center for Neurodegenerative Disease and MemoryImpairment, Capital Medical University, Beijing, P.R. China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.,National Clinical Research Center forGeriatric Disorders, Beijing, P.R. China
| | - Tingting Hou
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, P.R. China
| | - Yuanruhua Tian
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, P.R. China
| | - Qiaoqi Wu
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, P.R. China.,Clinical Center for Neurodegenerative Disease and MemoryImpairment, Capital Medical University, Beijing, P.R. China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.,National Clinical Research Center forGeriatric Disorders, Beijing, P.R. China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, P.R. China.,Clinical Center for Neurodegenerative Disease and MemoryImpairment, Capital Medical University, Beijing, P.R. China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.,National Clinical Research Center forGeriatric Disorders, Beijing, P.R. China
| | - Yiping Wei
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, P.R. China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, P.R. China.,Clinical Center for Neurodegenerative Disease and MemoryImpairment, Capital Medical University, Beijing, P.R. China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.,National Clinical Research Center forGeriatric Disorders, Beijing, P.R. China
| | - Xiu Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuan Wu Hospital, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
34
|
Zhu J, Xu S, Gao W, Feng J, Zhao G. Honokiol induces endoplasmic reticulum stress-mediated apoptosis in human lung cancer cells. Life Sci 2019; 221:204-211. [DOI: 10.1016/j.lfs.2019.01.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 11/15/2022]
|
35
|
Yi X, Guo W, Shi Q, Yang Y, Zhang W, Chen X, Kang P, Chen J, Cui T, Ma J, Wang H, Guo S, Chang Y, Liu L, Jian Z, Wang L, Xiao Q, Li S, Gao T, Li C. SIRT3-Dependent Mitochondrial Dynamics Remodeling Contributes to Oxidative Stress-Induced Melanocyte Degeneration in Vitiligo. Am J Cancer Res 2019; 9:1614-1633. [PMID: 31037127 PMCID: PMC6485185 DOI: 10.7150/thno.30398] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/22/2019] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial dysregulation has been implicated in oxidative stress-induced melanocyte destruction in vitiligo. However, the molecular mechanism underlying this process is merely investigated. Given the prominent role of nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase Sirtuin3 (SIRT3) in sustaining mitochondrial dynamics and homeostasis and that SIRT3 expression and activity can be influenced by oxidative stress-related signaling, we wondered whether SIRT3 could play an important role in vitiligo melanocyte degeneration by regulating mitochondrial dynamics. Methods: We initially testified SIRT3 expression and activity in normal and vitiligo melanocytes via PCR, immunoblotting and immunofluorescence assays. Then, cell apoptosis, mitochondrial function and mitochondrial dynamics after SIRT3 intervention were analyzed by flow cytometry, immunoblotting, confocal laser microscopy, transmission electron microscopy and oxphos activity assays. Chromatin immunoprecipitation (ChIP), co-immunoprecipitation (Co-IP), immunoblotting and immunofluorescence assays were performed to clarify the upstream regulatory mechanism of SIRT3. Finally, the effect of honokiol on protecting melanocytes and the underlying mechanism were investigated via flow cytometry and immunoblotting analysis. Results: We first found that the expression and the activity of SIRT3 were significantly impaired in vitiligo melanocytes both in vitro and in vivo. Then, SIRT3 deficiency led to more melanocyte apoptosis by inducing severe mitochondrial dysfunction and cytochrome c release to cytoplasm, with Optic atrophy 1 (OPA1)-mediated mitochondrial dynamics remodeling involved in. Moreover, potentiated carbonylation and dampened peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) activation accounted for SIRT3 dysregulation in vitiligo melanocytes. Finally, we proved that honokiol could prevent melanocyte apoptosis under oxidative stress by activating SIRT3-OPA1 axis. Conclusions: Overall, we demonstrate that SIRT3-dependent mitochondrial dynamics remodeling contributes to oxidative stress-induced melanocyte degeneration in vitiligo, and honokiol is promising in preventing oxidative stress-induced vitiligo melanocyte apoptosis.
Collapse
|
36
|
Chen H, Fu W, Chen H, You S, Liu X, Yang Y, Wei Y, Huang J, Rui W. Magnolol attenuates the inflammation and enhances phagocytosis through the activation of MAPK, NF-κB signal pathways in vitro and in vivo. Mol Immunol 2019; 105:96-106. [DOI: 10.1016/j.molimm.2018.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/22/2018] [Accepted: 11/11/2018] [Indexed: 12/25/2022]
|
37
|
Wang D, Dong X, Wang C. Honokiol Ameliorates Amyloidosis and Neuroinflammation and Improves Cognitive Impairment in Alzheimer's Disease Transgenic Mice. J Pharmacol Exp Ther 2018; 366:470-478. [PMID: 29991481 DOI: 10.1124/jpet.118.248674] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 07/05/2018] [Indexed: 03/08/2025] Open
Abstract
The present study examined the effects of honokiol on amyloid-β (Aβ)-induced cognitive impairment and the underlying mechanisms in APPswe/PS1dE9 transgenic mice. The results showed that honokiol administration (20 mg/kg per day, intraperitoneally) for 6 weeks effectively improved spatial memory deficits in APPswe/PS1dE9 transgenic mice. Honokiol significantly lowered Aβ production and senile plaque deposition by downregulating β-site amyloid precursor protein cleavage enzyme 1 and enhancing Aβ phagocytosis by microglia. Honokiol reduced glial cell activation and the production of proinflammatory cytokines (TNF-α, IL-1β, and IL-6). Honokiol increased the transcriptional activity and protein levels of peroxisome proliferator-activated receptor-γ (PPARγ) However, all of the beneficial effects of honokiol on pathologic changes, including biochemistry and cognitive function, could be blocked by GW9662, a specific PPARγ inhibitor. These findings suggested that honokiol may be a natural PPARγ agonist, acting to attenuate Aβ generation and neuroinflammation. Therefore, honokiol may be a potential therapeutic approach for Alzheimer's disease.
Collapse
Affiliation(s)
- Dongmei Wang
- Department of Pathogen Biology, Medical College, Henan University of Science and Technology, Luolong District, Luoyang, China
| | - Xiaohui Dong
- Department of Pathogen Biology, Medical College, Henan University of Science and Technology, Luolong District, Luoyang, China
| | - Chenying Wang
- Department of Pathogen Biology, Medical College, Henan University of Science and Technology, Luolong District, Luoyang, China
| |
Collapse
|
38
|
Neuroprotective effect of crocin on substantia nigra in MPTP-induced Parkinson's disease model of mice. Anat Sci Int 2018; 94:119-127. [PMID: 30159851 DOI: 10.1007/s12565-018-0457-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/16/2018] [Indexed: 02/07/2023]
Abstract
Parkinson's disease is caused by damage to substantia nigra dopaminergic neurons. Factors such as oxidative stress, inflammatory factors, and acetylcholinesterase activity may induce this disease. On the other hand, crocin-one of the active ingredients of saffron-has anti-oxidant and anti-inflammatory properties. This study was performed to evaluate the protective effect of crocin to decrease dopaminergic neuron damage and Parkinson's disease complications induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). A set of 24 male BALB/c Mice were divided randomly into four groups: (1) MPTP group receiving 30 mg/kg MPTP for 5 days; (2) MPTP + crocin group receiving 30 mg/kg MPTP for 5 days and 30 mg/kg crocin for 15 days; (3) NS group receiving normal saline for 5 days; and (4) NSIG group receiving normal saline intraperitoneally for 5 days and also normal saline by gavage for 15 days. After the treatment period, pole and hanging motor tests were performed in all groups. Then, the brains of all the animals were removed and fixed in formalin, prepared according to routine histologic methods and cut into sections of 5 µm thickness. Prepared sections were stained by immunohistochemistry techniques and toluidine blue to detect tyrosine-hydroxylase (TH)-positive neurons and dark neurons, respectively. Finally, the mean number of these cells were calculated by stereological methods and compared with the statistical tests in different groups. The results showed a significant increase in the time taken for the animal to fall from the pole in the MPTP group in comparison with other groups (P < 0.001). The time taken for them to stay on the wire in the hanging test decreased significantly in the MPTP group compared to the other groups (P < 0.001).,while in the MPTP + crocin group, the time to falling decreased (P < 0.05) and the time staying on the wire increased (P < 0.001) compared to the MPTP group. The number of TH-positive neurons in the MPTP group also decreased significantly in comparison with saline and MPTP + crocin groups (P < 0.001). The number of dark neuron sin the MPTP group increased significantly as compared with saline and the MPTP + Crocin groups (P < 0.001). Our results showed that crocin improves MPTP-induced Parkinson's disease complications and decreases cell death in the substantia nigra.
Collapse
|
39
|
Honokiol Protects against Anti- β1-Adrenergic Receptor Autoantibody-Induced Myocardial Dysfunction via Activation of Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1640804. [PMID: 30116474 PMCID: PMC6079338 DOI: 10.1155/2018/1640804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/04/2018] [Accepted: 05/30/2018] [Indexed: 12/19/2022]
Abstract
Myocardial diseases are prevalent syndromes with high mortality rate. The exploration of effective interference is important. Anti-β1-adrenergic receptor autoantibody (β1-AAB) is highly correlated with myocardial dysfunction. The actions and underlying mechanisms of honokiol (HNK) in β1-AAB-positive patients await to be unraveled. In this study, we established a rat model of β1-AAB positive with myocardial dysfunction. Cardiac function following β1-AR-ECII administration was analyzed using the VisualSonics Vevo 770 High-Resolution In Vivo Imaging System. The levels of autophagy-related proteins were detected by Western blotting. Our data revealed that HNK reversed β1-AAB-induced effects and protected myocardial tissues from dysfunction. After HNK treatment, the cardiac contractile ability increased and the LDH activity decreased. HNK attenuated myocardial degeneration. In addition, HNK promoted the activation of the AMP-dependent protein kinase/Unc-51-like autophagy activating kinase (AMPK/ULK) pathway and activated autophagy. These results suggest that HNK protects against β1-AAB-induced myocardial dysfunction via activation of autophagy and it may be a potentially therapeutic compound for β1-AAB-positive myocardial diseases.
Collapse
|
40
|
Gray NE, Zweig JA, Caruso M, Martin MD, Zhu JY, Quinn JF, Soumyanath A. Centella asiatica increases hippocampal synaptic density and improves memory and executive function in aged mice. Brain Behav 2018; 8:e01024. [PMID: 29920983 PMCID: PMC6043711 DOI: 10.1002/brb3.1024] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Centella asiatica is a plant used for centuries to enhance memory. We have previously shown that a water extract of Centella asiatica (CAW) attenuates age-related spatial memory deficits in mice and improves neuronal health. Yet the effect of CAW on other cognitive domains remains unexplored as does its mechanism of improving age-related cognitive impairment. This study investigates the effects of CAW on a variety of cognitive tasks as well as on synaptic density and mitochondrial and antioxidant pathways. METHODS Twenty-month-old CB6F1 mice were treated with CAW (2 mg/ml) in their drinking water for 2 weeks prior to behavioral testing. Learning, memory, and executive function were assessed using the novel object recognition task (NORT), object location memory task (OLM), and odor discrimination reversal learning (ODRL) test. Tissue was collected for Golgi analysis of spine density as well as assessment of mitochondrial, antioxidant, and synaptic proteins. RESULTS CAW improved performance in all behavioral tests suggesting effects on hippocampal and cortical dependent memory as well as on prefrontal cortex mediated executive function. There was also an increase in synaptic density in the treated animals, which was accompanied by increased expression of the antioxidant response gene NRF2 as well as the mitochondrial marker porin. CONCLUSIONS These data show that CAW can increase synaptic density as well as antioxidant and mitochondrial proteins and improve multiple facets of age-related cognitive impairment. Because mitochondrial dysfunction and oxidative stress also accompany cognitive impairment in many pathological conditions this suggests a broad therapeutic utility of CAW.
Collapse
Affiliation(s)
- Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | - Jonathan A Zweig
- Department of Neurology, Oregon Health and Science University, Portland, Oregon.,Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Maya Caruso
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | - Marjoen D Martin
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | - Jennifer Y Zhu
- Department of Neurology, Oregon Health and Science University, Portland, Oregon.,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, Oregon.,Department of Neurology and Parkinson's Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, Oregon
| | - Amala Soumyanath
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
41
|
Sahoo AK, Dandapat J, Dash UC, Kanhar S. Features and outcomes of drugs for combination therapy as multi-targets strategy to combat Alzheimer's disease. JOURNAL OF ETHNOPHARMACOLOGY 2018; 215:42-73. [PMID: 29248451 DOI: 10.1016/j.jep.2017.12.015] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD), a deleterious neurodegenerative disorder that impairs memory, cognitive functions and may lead to dementia in late stage of life. The pathogenic cause of AD remains incompletely understood and FDA approved drugs are partial inhibitors rather than curative. Most of drugs are synthetic or natural products as galanthamine is an alkaloid obtained from Galanthus spp. Huperzine A, an alkaloid found in Huperzia spp., gingkolides a diterpenoids from Gingko biloba and many ethnobotanicals like Withania somnifera (L.) Dunal., Physostigma venenosum Balf., Bacopa monnieri (L.) Wettst., Centella asiatica (L.) Urb. have been used by traditional Indian, Chinese, and European system of medicines in AD. Clinical significance opioid alkaloid in Papaver somniferum has shown another dimension to this study. Over exploitation of medicinal plants with limited bioactive principles has provided templates to design synthetic drugs in AD e.g. rivastigmine, phenserine, eptastigmine based on chemical structure of physostigmine of Physostigma venenosum Balf. Even ZT-1 a prodrug of Hup A and memogain a prodrug of galantamine has achieved new direction in drug development in AD. All these first-line cholinesterase-inhibitors are used as symptomatic treatments in AD. Single modality of "One-molecule-one-target" strategy for treating AD has failed and so future therapies on "Combination-drugs-multi-targets" strategy (CDMT) will need to address multiple aspects to block the progression of pathogenesis of AD. Besides, cholinergic and amyloid drugs, in this article we summarize proteinopathy-based drugs as AD therapeutics from a variety of biological sources. In this review, an attempt has been made to elucidate the molecular mode of action of various plant products, and synthetic drugs investigated in various preclinical and clinical tests in AD. It also discusses current attempts to formulate a comprehensive CDMT strategy to counter complex pathogenesis in AD. MATERIALS AND METHODS Information were collected from classical books on medicinal plants, pharmacopoeias and scientific databases like PubMed, Scopus, GoogleScholar, Web of Science and electronic searches were performed using Cochrane Library, Medline and EMBASE. Also published scientific literatures from Elsevier, Taylor and Francis, Springer, ACS, Wiley publishers and reports by government bodies and documentations were assessed. RESULTS 60 no. of natural and synthetic drugs have been studied with their significant bioactivities. A decision matrix designed for evaluation of drugs for considering to the hypothetic "CDMT" strategy in AD. We have introduced the scoring pattern of individual drugs and based on scoring pattern, drugs that fall within the scoring range of 18-25 are considered in the proposed CDMT. It also highlights the importance of available natural products and in future those drugs may be considered in CDMT along with the qualified synthetic drugs. CONCLUSION A successful validation of the CDMT strategy may open up a debate on health care reform to explore other possibilities of combination therapy. In doing so, it should focus on clinical and molecular relationships between AD and CDMT. A better understanding of these relationships could inform and impact future development of AD-directed treatment strategies. This strategy also involves in reducing costs in treatment phases which will be affordable to a common man suffering from AD.
Collapse
Affiliation(s)
- Atish Kumar Sahoo
- Phytotherapy Research Lab., Medicinal & Aromatic Plant Division, Regional Plant Resource Centre, Forest & Environment Department, Govt. of Odisha, Nayapalli, Bhubaneswar 751015, India.
| | - Jagnehswar Dandapat
- Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar 751004, India
| | - Umesh Chandra Dash
- Phytotherapy Research Lab., Medicinal & Aromatic Plant Division, Regional Plant Resource Centre, Forest & Environment Department, Govt. of Odisha, Nayapalli, Bhubaneswar 751015, India
| | - Satish Kanhar
- Phytotherapy Research Lab., Medicinal & Aromatic Plant Division, Regional Plant Resource Centre, Forest & Environment Department, Govt. of Odisha, Nayapalli, Bhubaneswar 751015, India
| |
Collapse
|
42
|
Sheng A, Zhang Y, Li G, Zhang G. Inhibitory Effects of Honokiol on the Voltage-Gated Potassium Channels in Freshly Isolated Mouse Dorsal Root Ganglion Neurons. Neurochem Res 2017; 43:450-457. [PMID: 29177805 DOI: 10.1007/s11064-017-2440-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/17/2017] [Accepted: 11/22/2017] [Indexed: 01/06/2023]
Abstract
Voltage-gated potassium (KV) currents, subdivided into rapidly inactivating A-type currents (I A) and slowly inactivating delayed rectifier currents (I K), play a fundamental role in modulating pain by controlling neuronal excitability. The effects of Honokiol (Hon), a natural biphenolic compound derived from Magnolia officinalis, on KV currents were investigated in freshly isolated mouse dorsal root ganglion neurons using the whole-cell patch clamp technique. Results showed that Hon inhibited I A and I K in concentration-dependent manner. The IC50 values for block of I A and I K were 30.5 and 25.7 µM, respectively. Hon (30 µM) shifted the steady-state activation curves of I A and I K to positive potentials by 17.6 and 16.7 mV, whereas inactivation and recovery from the inactivated state of I A were unaffected. These results suggest that Hon preferentially interacts with the active states of the I A and I K channels, and has no effect on the resting state and inactivated state of the I A channel. Blockade on K+ channels by Hon may contribute to its antinociceptive effect, especially anti-inflammatory pain.
Collapse
Affiliation(s)
- Anqi Sheng
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yan Zhang
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Guang Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Guangqin Zhang
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
43
|
Talarek S, Listos J, Barreca D, Tellone E, Sureda A, Nabavi SF, Braidy N, Nabavi SM. Neuroprotective effects of honokiol: from chemistry to medicine. Biofactors 2017; 43:760-769. [PMID: 28817221 DOI: 10.1002/biof.1385] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/21/2017] [Accepted: 07/27/2017] [Indexed: 01/15/2023]
Abstract
The incidence of neurological disorders is growing in developed countries together with increased lifespan. Nowadays, there are still no effective treatments for neurodegenerative pathologies, which make necessary to search for new therapeutic agents. Natural products, most of them used in traditional medicine, are considered promising alternatives for the treatment of neurodegenerative diseases. Honokiol is a natural bioactive phenylpropanoid compound, belonging to the class of neolignan, found in notable amounts in the bark of Magnolia tree, and has been reported to exert diverse pharmacological properties including neuroprotective activities. Honokiol can permeate the blood brain barrier and the blood-cerebrospinal fluid to increase its bioavailability in neurological tissues. Diverse studies have provided evidence on the neuroprotective effect of honokiol in the central nervous system, due to its potent antioxidant activity, and amelioration of the excitotoxicity mainly related to the blockade of glutamate receptors and reduction in neuroinflammation. In addition, recent studies suggest that honokiol can attenuate neurotoxicity exerted by abnormally aggregated Aβ in Alzheimer's disease. The present work summarizes what is currently known concerning the neuroprotective effects of honokiol and its potential molecular mechanisms of action, which make it considered as a promising agent in the treatment and management of neurodegenerative diseases. © 2017 BioFactors, 43(6):760-769, 2017.
Collapse
Affiliation(s)
- Sylwia Talarek
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Lublin 20-093, Poland
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Lublin 20-093, Poland
| | - Davide Barreca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Ester Tellone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress (NUCOX) and CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), University of Balearic Islands, Balearic Islands, Spain
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Australia
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Kantham S, Chan S, McColl G, Miles JA, Veliyath SK, Deora GS, Dighe SN, Khabbazi S, Parat MO, Ross BP. Effect of the Biphenyl Neolignan Honokiol on Aβ 42-Induced Toxicity in Caenorhabditis elegans, Aβ 42 Fibrillation, Cholinesterase Activity, DPPH Radicals, and Iron(II) Chelation. ACS Chem Neurosci 2017. [PMID: 28650631 DOI: 10.1021/acschemneuro.7b00071] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The biphenyl neolignan honokiol is a neuroprotectant which has been proposed as a treatment for central nervous system disorders such as Alzheimer's disease (AD). The death of cholinergic neurons in AD is attributed to multiple factors, including accumulation and fibrillation of amyloid beta peptide (Aβ) within the brain; metal ion toxicity; and oxidative stress. In this study, we used a transgenic Caenorhabditis elegans model expressing full length Aβ42 as a convenient in vivo system for examining the effect of honokiol against Aβ-induced toxicity. Furthermore, honokiol was evaluated for its ability to inhibit Aβ42 oligomerization and fibrillation; inhibit acetylcholinesterase and butyrylcholinesterase; scavenge 2,2-diphenyl-1-picrylhydrazyl (DPPH) radicals; and chelate iron(II). Honokiol displayed activity similar to that of resveratrol and (-)-epigallocatechin gallate (EGCG) in delaying Aβ42-induced paralysis in C. elegans, and it exhibited moderate-to-weak ability to inhibit Aβ42 on-pathway aggregation, inhibit cholinesterases, scavenge DPPH radicals, and chelate iron(II). Moreover, honokiol was found to be chemically stable relative to EGCG, which was highly unstable. Together with its good drug-likeness and brain availability, these results suggest that honokiol may be amenable to drug development and that the synthesis of honokiol analogues to optimize these properties should be considered.
Collapse
Affiliation(s)
- Srinivas Kantham
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Stephen Chan
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Gawain McColl
- The
Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jared A. Miles
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Suresh Kumar Veliyath
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Girdhar Singh Deora
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Satish N. Dighe
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Samira Khabbazi
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Marie-Odile Parat
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Benjamin P. Ross
- School
of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
45
|
Quantitative analysis of the therapeutic effect of magnolol on MPTP-induced mouse model of Parkinson's disease using in vivo 18F-9-fluoropropyl-(+)-dihydrotetrabenazine PET imaging. PLoS One 2017; 12:e0173503. [PMID: 28257461 PMCID: PMC5336287 DOI: 10.1371/journal.pone.0173503] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/21/2017] [Indexed: 02/04/2023] Open
Abstract
18F-9-Fluoropropyl-(+)-dihydrotetrabenazine [18F-FP-(+)-DTBZ] positron emission tomography (PET) has been shown to detect dopaminergic neuron loss associated with Parkinson’s disease (PD) in human and neurotoxin-induced animal models. A polyphenol compound, magnolol, was recently proposed as having a potentially restorative effect in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)- or 6-hydroxydopamine-treated animal models. In this study, 18F-FP-(+)-DTBZ PET was used to determine the therapeutic efficacy of magnolol in an MPTP–PD mouse model that was prepared by giving an intraperitoneally (i.p.) daily dose of 25 mg/kg MPTP to male C57BL/6 mice for 5 consecutive days. Twenty-minute static 18F-FP-(+)-DTBZ PET scans were performed before MPTP treatment and 5 days after the termination of MPTP treatment to set up the baseline control. Half of the MPTP-treated mice then received a daily dose of magnolol (10 mg/kg dissolved in corn oil, i.p.) for 6 days. 18F-FP-(+)-DTBZ PET imaging was performed the day after the final treatment. All 18F-FP-(+)-DTBZ PET images were analysed and the specific uptake ratio (SUr) was calculated. Ex vivo autoradiography (ARG) and corresponding immunohistochemistry (IHC) studies were conducted to confirm the distribution of dopaminergic terminals in the striatum. The striatal SUr ratios of 18F-FP-(+)-DTBZ PET images for the Sham, the MPTP, and the MPTP + Magnolol-treated groups were 1.25 ± 0.05, 0.75 ± 0.06, and 1.00 ± 0.11, respectively (n = 4 for each group). The ex vivo 18F-FP-(+)-DTBZ ARG and IHC results correlated favourably with the PET imaging results. 18F-FP-(+)-DTBZ PET imaging suggested that magnolol post-treatment may reverse the neuronal damage in the MPTP-lesioned PD mice. In vivo imaging of the striatal vesicular monoamine transporter type 2 (VMAT2) distribution using 18F-FP-(+)-DTBZ animal PET is a useful method to evaluate the efficacy of therapeutic drugs i.e., magnolol, for the management of PD.
Collapse
|
46
|
Development of a Prolonged-Release Drug Delivery System with Magnolol Loaded in Amino-Functionalized Mesoporous Silica. APPLIED SCIENCES-BASEL 2017. [DOI: 10.3390/app7030237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
47
|
Matsui N, Akae H, Hirashima N, Kido Y, Tanabe S, Koseki M, Fukuyama Y, Akagi M. Magnolol Enhances Hippocampal Neurogenesis and Exerts Antidepressant-Like Effects in Olfactory Bulbectomized Mice. Phytother Res 2016; 30:1856-1861. [DOI: 10.1002/ptr.5695] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/06/2016] [Accepted: 07/20/2016] [Indexed: 01/08/2023]
Affiliation(s)
- Nobuaki Matsui
- Faculty of Pharmaceutical Sciences; Tokushima Bunri University; 180 Nishihama-bouji Yamashiro-cho Tokushima 770-8514 Japan
| | - Haruka Akae
- Faculty of Pharmaceutical Sciences; Tokushima Bunri University; 180 Nishihama-bouji Yamashiro-cho Tokushima 770-8514 Japan
| | - Nana Hirashima
- Faculty of Pharmaceutical Sciences; Tokushima Bunri University; 180 Nishihama-bouji Yamashiro-cho Tokushima 770-8514 Japan
| | - Yuki Kido
- Faculty of Pharmaceutical Sciences; Tokushima Bunri University; 180 Nishihama-bouji Yamashiro-cho Tokushima 770-8514 Japan
| | - Satoshi Tanabe
- Faculty of Pharmaceutical Sciences; Tokushima Bunri University; 180 Nishihama-bouji Yamashiro-cho Tokushima 770-8514 Japan
| | - Mayumi Koseki
- Faculty of Pharmaceutical Sciences; Tokushima Bunri University; 180 Nishihama-bouji Yamashiro-cho Tokushima 770-8514 Japan
| | - Yoshiyasu Fukuyama
- Faculty of Pharmaceutical Sciences; Tokushima Bunri University; 180 Nishihama-bouji Yamashiro-cho Tokushima 770-8514 Japan
| | - Masaaki Akagi
- Faculty of Pharmaceutical Sciences; Tokushima Bunri University; 180 Nishihama-bouji Yamashiro-cho Tokushima 770-8514 Japan
| |
Collapse
|
48
|
Neuroprotective effects of honokiol against beta-amyloid-induced neurotoxicity via GSK-3β and β-catenin signaling pathway in PC12 cells. Neurochem Int 2016; 97:8-14. [DOI: 10.1016/j.neuint.2016.04.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 11/23/2022]
|
49
|
Comparative metabolism of honokiol in mouse, rat, dog, monkey, and human hepatocytes. Arch Pharm Res 2016; 39:516-530. [PMID: 26983827 DOI: 10.1007/s12272-016-0731-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/08/2016] [Indexed: 10/22/2022]
Abstract
Honokiol has antitumor, antioxidative, anti-inflammatory, and antithrombotic effects. Here we aimed to identify the metabolic profile of honokiol in mouse, rat, dog, monkey, and human hepatocytes and to characterize the enzymes responsible for the glucuronidation and sulfation of honokiol. Honokiol had a high hepatic extraction ratio in all five species, indicating that it was extensively metabolized. A total of 32 metabolites, including 17 common and 15 different metabolites, produced via glucuronidation, sulfation, and oxidation of honokiol allyl groups were tentatively identified using liquid chromatography-high resolution quadrupole Orbitrap mass spectrometry. Glucuronidation of honokiol to M8 (honokiol-4-glucuronide) and M9 (honokiol-2'-glucuronide) was the predominant metabolic pathway in hepatocytes of all five species; however, interspecies differences between 4- and 2'-glucuronidation of honokiol were observed. UGT1A1, 1A8, 1A9, 2B15, and 2B17 played major roles in M8 formation, whereas UGT1A7 and 1A9 played major roles in M9 formation. Human cDNA-expressed SULT1C4 played a major role in M10 formation (honokiol-2'-sulfate), whereas SULT1A1*1, 1A1*2, and 1A2 played major roles in M11 formation (honokiol-4-sulfate). In conclusion, honokiol metabolism showed interspecies differences.
Collapse
|
50
|
Nakai M, Iizuka M, Matsui N, Hosogi K, Imai A, Abe N, Shiraishi H, Hirata A, Yagi Y, Jobu K, Yokota J, Kato E, Hosoda S, Yoshioka S, Harada K, Kubo M, Fukuyama Y, Miyamura M. Bangle (Zingiber purpureum) Improves Spatial Learning, Reduces Deficits in Memory, and Promotes Neurogenesis in the Dentate Gyrus of Senescence-Accelerated Mouse P8. J Med Food 2016; 19:435-41. [PMID: 26829513 DOI: 10.1089/jmf.2015.3562] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Bangle (Zingiber purpureum) is a tropical ginger that is used as a spice in Southeast Asia. Phenylbutenoid dimers isolated from Bangle have exhibited neurotrophic effects in primary cultured rat cortical neurons and PC12 cells. Furthermore, chronic treatment with phenylbutenoid dimers enhances hippocampal neurogenesis in olfactory bulbectomized mice. In this study, we investigated the effects of Bangle extract on behavior and hippocampal neurogenesis in vivo. SAMP8 mice, which are an established model for accelerated aging, with age-related learning and memory impairments, were given a Bangle-containing diet for 1 month, and subsequent behavioral tests and immunohistochemistry for Ki67, a proliferating cell marker, were performed. We found that the Bangle-containing diet improved spatial learning and memory deficits in the Morris water maze and significantly increased the numbers of Ki67-positive cells in the dentate gyrus of the SAMP8 mice. In addition, the Bangle extract exhibited a neurotrophin-like activity as indicated by the induction of neurite sprouting in PC12 cells. Our results suggest that Bangle is beneficial for the prevention of age-related progression of cognitive impairment.
Collapse
Affiliation(s)
- Megumi Nakai
- 1 Department of Pharmacy, Kochi Medical Graduate School , Kochi, Japan .,2 Department of Pharmacy, Kochi Medical School Hospital , Kochi, Japan
| | - Michiro Iizuka
- 2 Department of Pharmacy, Kochi Medical School Hospital , Kochi, Japan
| | - Nobuaki Matsui
- 3 Faculty of Pharmaceutical Sciences, Tokushima Bunri University , Tokushima, Japan
| | - Kazuko Hosogi
- 1 Department of Pharmacy, Kochi Medical Graduate School , Kochi, Japan
| | - Akiko Imai
- 2 Department of Pharmacy, Kochi Medical School Hospital , Kochi, Japan
| | - Noriaki Abe
- 1 Department of Pharmacy, Kochi Medical Graduate School , Kochi, Japan .,2 Department of Pharmacy, Kochi Medical School Hospital , Kochi, Japan
| | - Hisashi Shiraishi
- 1 Department of Pharmacy, Kochi Medical Graduate School , Kochi, Japan .,2 Department of Pharmacy, Kochi Medical School Hospital , Kochi, Japan
| | - Ayumu Hirata
- 1 Department of Pharmacy, Kochi Medical Graduate School , Kochi, Japan .,2 Department of Pharmacy, Kochi Medical School Hospital , Kochi, Japan
| | - Yusuke Yagi
- 1 Department of Pharmacy, Kochi Medical Graduate School , Kochi, Japan .,2 Department of Pharmacy, Kochi Medical School Hospital , Kochi, Japan
| | - Kohei Jobu
- 2 Department of Pharmacy, Kochi Medical School Hospital , Kochi, Japan
| | - Junko Yokota
- 2 Department of Pharmacy, Kochi Medical School Hospital , Kochi, Japan
| | | | | | - Saburo Yoshioka
- 3 Faculty of Pharmaceutical Sciences, Tokushima Bunri University , Tokushima, Japan
| | - Kenichi Harada
- 3 Faculty of Pharmaceutical Sciences, Tokushima Bunri University , Tokushima, Japan
| | - Miwa Kubo
- 3 Faculty of Pharmaceutical Sciences, Tokushima Bunri University , Tokushima, Japan
| | - Yoshiyasu Fukuyama
- 3 Faculty of Pharmaceutical Sciences, Tokushima Bunri University , Tokushima, Japan
| | - Mitsuhiko Miyamura
- 1 Department of Pharmacy, Kochi Medical Graduate School , Kochi, Japan .,2 Department of Pharmacy, Kochi Medical School Hospital , Kochi, Japan
| |
Collapse
|