1
|
Calabrese EJ, Pressman P, Hayes AW, Agathokleous E, Dhawan G, Kapoor R, Parmar J, Mssillou I, Calabrese V. Fisetin: hormesis accounts for many of its chemoprotective effects. Biogerontology 2025; 26:90. [PMID: 40208387 DOI: 10.1007/s10522-025-10230-1] [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: 02/12/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
The present paper provides the first integrated assessment of the capacity of the flavonol, fisetin, to induce hormetic dose responses. Fisetin was shown to induce hormetic dose responses in cellular and in vivo animal model systems affecting a broad range of endpoints of potential therapeutic and public health significance across the entire lifespan. Fisetin was effective in slowing aging processes, acting as a senolytic agent in multiple organ systems, in an hormetic fashion. In addition, fisetin was broadly neuroprotective, including during fetal development, and preventing the toxicity of methylmercury. Since these findings indicate that fisetin may have the potential to induce multi-system chemoprotective effects, it indicates the need to better clarify the absorption and bioavailability of fisetin and ways to enhance its efficiency.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health, School of Public Health and Health Sciences, University of Massachusetts, Morrill I-N344, Amherst, MA, 01003, USA.
| | - Peter Pressman
- University of Maine, 5728 Fernald Hall, Room 201, Orono, ME, 04469, USA
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Evgenios Agathokleous
- School of Ecology and Applied Meteorology, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Gaurav Dhawan
- Sri Guru Ram Das (SGRD) University of Health Sciences Amritsar, India, Hartford, CT, United States
| | - Rachna Kapoor
- Saint Francis Hospital and Medical Center, Hartford, CT, USA
| | | | - Ibrahim Mssillou
- National Agency of Medicinal and Aromatic Plants, BP 159, Principal, 34000, Taounate, Morocco
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via Santa Sofia 97, 95123, Catania, Italy
| |
Collapse
|
2
|
Maher P, Christopher R, Evans R, Raschke W. Safety, Pharmacokinetics, and Cardiodynamics of CMS121, a Novel Small Molecule Fisetin Derivative with Neuroprotective Properties, in Phase 1 Healthy Adult Volunteers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.28.25323123. [PMID: 40093210 PMCID: PMC11908328 DOI: 10.1101/2025.02.28.25323123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The safety, tolerability, pharmacokinetics, age-related effects of single (SD) and repeat (RD) doses of CMS121, a novel small molecule fisetin derivative, were evaluated in healthy adult volunteers. The effects of food were also evaluated in healthy young adult subjects. SD of up to 1800 mg or RD up to 900 mg/day for 7 days was generally well tolerated, with the majority of TEAEs mild in severity. Generally, the pharmacokinetics of CMS121 and its metabolites were well characterized and increased in a dose-proportional or slightly greater than dose-proportional manner across the range of doses assessed. CMS121-C2 metabolite appears to contribute the most to the presence of the molar-equivalent CMS121 in plasma than the parent compound or the other metabolites (i.e. CMS121-C1 and CMS121-C3). Urinary excretion of CMS121 metabolites was minimal, implying urinary excretion may not be a major clearance route by which CMS121 is eliminated after oral dosing. There is a significant effect of age on the pharmacokinetics of CMS121 and its metabolites, with higher systemic exposures to CMS121 and its metabolites and longer terminal elimination half-lives in elderly subjects. Systemic exposures to CMS121 were higher in the fed state by approximately 50%.
Collapse
|
3
|
Hirata Y, Nagase H, Satoh K, Takemori H, Furuta K, Kamatari YO. Antiferroptotic properties of allicin and related organosulfur compounds-diallyl disulfide and diallyl trisulfide-from Garlic. Food Chem Toxicol 2025; 195:115124. [PMID: 39580015 DOI: 10.1016/j.fct.2024.115124] [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: 09/27/2024] [Revised: 11/08/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Allicin (diallyl thiosulfinate) is an abundant bioactive compound in garlic (Allium sativum L.) with broad-spectrum antiinflammatory, antifungal, antioxidant, and antitumor properties. The bioactive compounds of garlic including allicin may also help reduce the incidence of various diseases, although the individual contributions and precise mechanisms are still largely unknown. In this study, we reveal that allicin and the closely related compounds diallyl disulfide and diallyl trisulfide (all containing more than one disulfide bond) protect mouse hippocampal HT22 cells against chemically induced ferroptosis, a newly defined form of cell death characterized by iron-dependent lipid peroxidation. In contrast, these organosulfur compounds did not prevent chemically induced apoptosis. The antiferroptotic efficacies of these compounds were strongly associated with prevention of lipid peroxidation and reactive oxygen species production but independent of glutathione upregulation, ferrous iron chelation, and modulation of the nuclear factor erythroid 2 (NF-E2)-related factor-2-antioxidant element response pathway. Additional studies are warranted on the therapeutic potential of allicin and related compounds in garlic for neurodegenerative diseases associated with ferroptosis.
Collapse
Affiliation(s)
- Yoko Hirata
- Life Science Research Center, Institute for Advanced Study, Gifu University, Gifu, 501-1193, Japan.
| | - Haruna Nagase
- Department of Pharmacology, Asahi University School of Dentistry, Gifu 501-0296, Japan
| | - Keitaro Satoh
- Department of Pharmacology, Asahi University School of Dentistry, Gifu 501-0296, Japan
| | - Hiroshi Takemori
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Gifu, 501-1193, Japan; Graduate School of Natural Science and Technology, Gifu University, Gifu, 501-1193, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, 501-1193, Japan
| | | | - Yuji O Kamatari
- Life Science Research Center, Institute for Advanced Study, Gifu University, Gifu, 501-1193, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, 501-1193, Japan; Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| |
Collapse
|
4
|
Balaji PG, Bhimrao LS, Yadav AK. Revolutionizing Stroke Care: Nanotechnology-Based Brain Delivery as a Novel Paradigm for Treatment and Diagnosis. Mol Neurobiol 2025; 62:184-220. [PMID: 38829514 DOI: 10.1007/s12035-024-04215-3] [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: 03/19/2024] [Accepted: 05/03/2024] [Indexed: 06/05/2024]
Abstract
Stroke, a severe medical condition arising from abnormalities in the coagulation-fibrinolysis cycle and metabolic processes, results in brain cell impairment and injury due to blood flow obstruction within the brain. Prompt and efficient therapeutic approaches are imperative to control and preserve brain functions. Conventional stroke medications, including fibrinolytic agents, play a crucial role in facilitating reperfusion to the ischemic brain. However, their clinical efficacy is hampered by short plasma half-lives, limited brain tissue distribution attributed to the blood-brain barrier (BBB), and lack of targeted drug delivery to the ischemic region. To address these challenges, diverse nanomedicine strategies, such as vesicular systems, polymeric nanoparticles, dendrimers, exosomes, inorganic nanoparticles, and biomimetic nanoparticles, have emerged. These platforms enhance drug pharmacokinetics by facilitating targeted drug accumulation at the ischemic site. By leveraging nanocarriers, engineered drug delivery systems hold the potential to overcome challenges associated with conventional stroke medications. This comprehensive review explores the pathophysiological mechanism underlying stroke and BBB disruption in stroke. Additionally, this review investigates the utilization of nanocarriers for current therapeutic and diagnostic interventions in stroke management. By addressing these aspects, the review aims to provide insight into potential strategies for improving stroke treatment and diagnosis through a nanomedicine approach.
Collapse
Affiliation(s)
- Paul Gajanan Balaji
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli (An Institute of National Importance under Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India
| | - Londhe Sachin Bhimrao
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli (An Institute of National Importance under Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India
| | - Awesh K Yadav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli (An Institute of National Importance under Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India.
| |
Collapse
|
5
|
Goujon M, Liang Z, Soriano-Castell D, Currais A, Maher P. The Neuroprotective Flavonoids Sterubin and Fisetin Maintain Mitochondrial Health under Oxytotic/Ferroptotic Stress and Improve Bioenergetic Efficiency in HT22 Neuronal Cells. Antioxidants (Basel) 2024; 13:460. [PMID: 38671908 PMCID: PMC11047672 DOI: 10.3390/antiox13040460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The global increase in the aging population has led to a rise in many age-related diseases with continuing unmet therapeutic needs. Research into the molecular mechanisms underlying both aging and neurodegeneration has identified promising therapeutic targets, such as the oxytosis/ferroptosis cell death pathway, in which mitochondrial dysfunction plays a critical role. This study focused on sterubin and fisetin, two flavonoids from the natural pharmacopeia previously identified as strong inhibitors of the oxytosis/ferroptosis pathway. Here, we investigated the effects of the compounds on the mitochondrial physiology in HT22 hippocampal nerve cells under oxytotic/ferroptotic stress. We show that the compounds can restore mitochondrial homeostasis at the level of redox regulation, calcium uptake, biogenesis, fusion/fission dynamics, and modulation of respiration, leading to the enhancement of bioenergetic efficiency. However, mitochondria are not required for the neuroprotective effects of sterubin and fisetin, highlighting their diverse homeostatic impacts. Sterubin and fisetin, thus, provide opportunities to expand drug development strategies for anti-oxytotic/ferroptotic agents and offer new perspectives on the intricate interplay between mitochondrial function, cellular stress, and the pathophysiology of aging and age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Marie Goujon
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, San Diego, CA 92037, USA
| | - Zhibin Liang
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, San Diego, CA 92037, USA
| | - David Soriano-Castell
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, San Diego, CA 92037, USA
| | - Antonio Currais
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, San Diego, CA 92037, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, San Diego, CA 92037, USA
| |
Collapse
|
6
|
Gu X, Xie Y, Cao Q, Hou Z, Zhang Y, Wang W. Fisetin alleviates cerebral ischemia/reperfusion injury by regulating Sirt1/Foxc1/Ubqln1 pathway-mediated proteostasis. Int Immunopharmacol 2024; 130:111742. [PMID: 38452414 DOI: 10.1016/j.intimp.2024.111742] [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: 12/27/2022] [Revised: 02/02/2024] [Accepted: 02/20/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Cerebral ischemia/reperfusion injury (IRI) is pathologically associated with protein damage. The flavonoid fisetin has good therapeutic effects on cerebral IRI. However, the role of fisetin in regulating protein damage during cerebral IRI development remains unclear. This study investigated the pharmacological effects of fisetin on protein damage during cerebral IRI progression and defined the underlying mechanism of action. METHODS In vivo and in vitro models of cerebral IRI were established by middle cerebral artery occlusion/reperfusion (MACO/R) and oxygen-glucose deprivation/reperfusion (OGD/R) treatment, respectively. Triphenyl tetrazolium chloride staining was performed to detect cerebral infarct size, and the modified neurologic severity score was used to examine neurological deficits. LDH activity and protein damage were assessed using kits. HT22 cell vitality and apoptosis were examined using CCK-8 assay and TUNEL staining, respectively. Interactions between Foxc1, Ubqln1, Sirt1, and Ezh2 were analyzed using CoIP, ChIP and/or dual-luciferase reporter gene assays. RESULTS Fisetin alleviated protein damage and ubiquitinated protein aggregation and neuronal death caused by MCAO/R and OGD/R. Ubqln1 knockdown abrogated the inhibitory effect of fisetin on OGD/R-induced protein damage, ubiquitinated protein aggregation, and neuronal death in HT22 cells. Further experiments demonstrated that Foxc1 functions as a transcriptional activator of Ubqln1 and that Sirt1 promotes Foxc1 expression by deacetylating Ezh2 and inhibiting its activity. Furthermore, Sirt1 knockdown abrogated fisetin-mediated biological effects on OGD/R-treated HT22 cells. CONCLUSION Fisetin improved proteostasis during cerebral IRI by regulating the Sirt1/Foxc1/Ubqln1 signaling axis. Our findings strongly suggest that fisetin-mediated inhibition of protein damage after ischemic stroke is a part of the mechanism through which fisetin is neuroprotective in cerebral IRI.
Collapse
Affiliation(s)
- Xunhu Gu
- Department of Neurology, The Second Affliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Yuqin Xie
- Department of Laboratory Medicine, Nanchang medical College, Nanchang 330006, Jiangxi, China
| | - Qian Cao
- Department of Neurology, The Second Affliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Zhuo Hou
- Department of Neurology, The Second Affliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Yan Zhang
- Department of Neurosurgery, The Second Affliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China.
| | - Wei Wang
- Department of Neurology, The Second Affliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China.
| |
Collapse
|
7
|
Adeli OA, Heidari-Soureshjani S, Rostamian S, Azadegan-Dehkordi Z, Khaghani A. Effects and Mechanisms of Fisetin against Ischemia-reperfusion Injuries: A Systematic Review. Curr Pharm Biotechnol 2024; 25:2138-2153. [PMID: 38310454 DOI: 10.2174/0113892010281821240102105415] [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: 09/12/2023] [Revised: 12/13/2023] [Accepted: 12/22/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) is a well-known ailment that can disturb organ function. OBJECTIVES This systematic review study investigated fisetin's effects and possible mechanisms in attenuating myocardial, cerebral, renal, and hepatic IRIs. METHODS This systematic review included studies earlier than Sep 2023 by following the PRISMA statement 2020. After determining inclusion and exclusion criteria and related keywords, bibliographic databases, such as Cochrane Library, PubMed, Web of Science, Embase, and Scopus databases, were used to search the relevant studies. Studies were imported in End- Note X8, and the primary information was recorded in Excel. RESULTS Fisetin reduced reactive oxygen species (ROS) generation and upregulated antioxidant enzymes, such as superoxide dismutase (SOD), glutathione (GSH), catalase (CAT), and glutathione peroxidase (GPx), in ischemic tissues. Moreover, fisetin can attenuate oxidative stress by activating phosphoinositide-3-kinase-protein kinase B/Akt (PI3K/Akt) and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathways. Fisetin has been indicated to prevent the activation of several pro-inflammatory signaling pathways, including NF-κB (Nuclear factor kappa-light-chain-enhancer of activated B cells) and MAPKs (Mitogen-activated protein kinases). It also inhibits the production of pro-inflammatory cytokines and enzymes like tumor necrosis factor-a (TNF-α), inducible-NO synthase (iNOS), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), interleukin-1β (IL-1β), IL-1, and IL-6. Fisetin attenuates IRI by improving mitochondrial function, anti-apoptotic effects, promoting autophagy, and preserving tissues from histological changes induced by IRIs. CONCLUSION Fisetin, by antioxidant, anti-inflammatory, mitochondrial protection, promoting autophagy, and anti-apoptotic properties, can reduce cell injury due to myocardial, cerebral renal, and hepatic IRIs without any significant side effects.
Collapse
Affiliation(s)
- Omid-Ali Adeli
- Department of Pathology, Lorestan University of Medical Sciences, Khorramabad, Iran
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | - Sahar Rostamian
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Zahra Azadegan-Dehkordi
- Oriented Nursing Midwifery Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Armin Khaghani
- Skin Diseases and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
8
|
Wei W, Jing L, Tian Y, Więckowska A, Kang D, Meng B, Panek D, Godyń J, Góral I, Song Y, Liu X, Zhan P. Multifunctional agents against Alzheimer's disease based on oxidative stress: Polysubstituted pyrazine derivatives synthesized by multicomponent reactions. Bioorg Med Chem 2023; 96:117535. [PMID: 37956505 DOI: 10.1016/j.bmc.2023.117535] [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: 09/12/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023]
Abstract
As Alzheimer's disease (AD) is a neurodegenerative disease with a complex pathogenesis, the exploration of multi-target drugs may be an effective strategy for AD treatment. Multifunctional small molecular agents can be obtained by connecting two or more active drugs or privileged pharmacophores by multicomponent reactions (MCRs). In this paper, two series of polysubstituted pyrazine derivatives with multifunctional moieties were designed as anti-AD agents and synthesized by Passerini-3CR and Ugi-4CR. Since the oxidative stress plays an important role in the pathological process of AD, the antioxidant activities of the newly synthesized compounds were first evaluated. Subsequently, selected active compounds were further screened in a series of AD-related bioassays, including Aβ1-42 self-aggregation and deaggregation, BACE-1 inhibition, metal chelation, and protection of SH-SY5Y cells from H2O2-induced oxidative damage. Compound A3B3C1 represented the best one with multifunctional potencies. Mechanism study showed that A3B3C1 acted on Nrf2/ARE signaling pathway, thus increasing the expression of related antioxidant proteins NQO1 and HO-1 to normal cell level. Furthermore, A3B3C1 showed good in vitro human plasma and liver microsome stability, indicating a potential for further development as multifunctional anti-AD agent.
Collapse
Affiliation(s)
- Wenxiu Wei
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Lanlan Jing
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Ye Tian
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China; Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| | - Anna Więckowska
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Bairu Meng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Dawid Panek
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Justyna Godyń
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Izabella Góral
- Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Yuning Song
- Department of Clinical Pharmacy, Qilu Hospital of Shandong University, Jinan, Shandong 250012, PR China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
9
|
Gong K, Zhang Z, Chen SS, Zhu XR, Wang MY, Yang XY, Ding C, Han JH, Li QS, Duan YJ. 6-Methyl flavone inhibits Nogo-B expression and improves high fructose diet-induced liver injury in mice. Acta Pharmacol Sin 2023; 44:2216-2229. [PMID: 37402997 PMCID: PMC10618526 DOI: 10.1038/s41401-023-01121-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023]
Abstract
Excessive fructose consumption increases hepatic de novo lipogenesis, resulting in cellular stress, inflammation and liver injury. Nogo-B is a resident protein of the endoplasmic reticulum that regulates its structure and function. Hepatic Nogo-B is a key protein in glycolipid metabolism, and inhibition of Nogo-B has protective effects against metabolic syndrome, thus small molecules that inhibit Nogo-B have therapeutic benefits for glycolipid metabolism disorders. In this study we tested 14 flavones/isoflavones in hepatocytes using dual luciferase reporter system based on the Nogo-B transcriptional response system, and found that 6-methyl flavone (6-MF) exerted the strongest inhibition on Nogo-B expression in hepatocytes with an IC50 value of 15.85 μM. Administration of 6-MF (50 mg· kg-1 ·d-1, i.g. for 3 weeks) significantly improved insulin resistance along with ameliorated liver injury and hypertriglyceridemia in high fructose diet-fed mice. In HepG2 cells cultured in a media containing an FA-fructose mixture, 6-MF (15 μM) significantly inhibited lipid synthesis, oxidative stress and inflammatory responses. Furthermore, we revealed that 6-MF inhibited Nogo-B/ChREBP-mediated fatty acid synthesis and reduced lipid accumulation in hepatocytes by restoring cellular autophagy and promoting fatty acid oxidation via the AMPKα-mTOR pathway. Thus, 6-MF may serve as a potential Nogo-B inhibitor to treat metabolic syndrome caused by glycolipid metabolism dysregulation.
Collapse
Affiliation(s)
- Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Zhen Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Sha-Sha Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Xin-Ran Zhu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Meng-Yao Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Xin-Yue Yang
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Chen Ding
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Ji-Hong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Qing-Shan Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China.
| | - Ya-Jun Duan
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
10
|
Huang Y, Dou Y, Yang B, He B, Zhang X, Zhang K, Yang X. Nicotinamide mononucleotide supplementation mitigates osteopenia induced by modeled microgravity in rats. Cell Stress Chaperones 2023; 28:385-394. [PMID: 37195399 PMCID: PMC10352228 DOI: 10.1007/s12192-023-01356-7] [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: 04/30/2023] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/18/2023] Open
Abstract
Exposure to weightlessness causes severe osteopenia, resulting in raised fracture risk. The current study aimed to investigate whether nicotinamide mononucleotide (NMN) supplementation protected against the osteopenia in hindlimb unloading (HLU) rats in vivo and modeled microgravity-induced osteoblastic dysfunction in vitro. The 3-mo-old rats were exposed to HLU and intragastrically administered NMN every 3 days (500 mg/kg body weight) for 4 weeks. NMN supplementation mitigated HLU-induced bone loss, evidenced by greater bone mass and biomechanical properties and better trabecular bone structure. NMN supplementation mitigated HLU-induced oxidative stress, evidenced by greater levels of nicotinamide adenine dinucleotide and activities of superoxide dismutase 2 and lesser malondialdehyde levels. Modeled microgravity stimulation using rotary wall vessel bioreactor in MC3T3-E1 cells inhibited osteoblast differentiation, which was reversed by NMN treatment. Furthermore, NMN treatment mitigated microgravity-induced mitochondrial impairments, evidenced by lesser reactive oxygen species generation and greater adenosine triphosphate production, mtDNA copy number, and activities of superoxide dismutase 2 and Complex I and II. Additionally, NMN promoted activation of AMP-activated protein kinase (AMPK), evidenced by greater AMPKα phosphorylation. Our research suggested that NMN supplementation attenuated osteoblastic mitochondrial impairment and mitigated osteopenia induced by modeled microgravity.
Collapse
Affiliation(s)
- Yunfei Huang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Yusheng Dou
- Department of Should and Elbow Joint, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Bo Yang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Baorong He
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Xuefang Zhang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Ke Zhang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Xiaobin Yang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China.
| |
Collapse
|
11
|
Choudhary N, Tewari D, Nabavi SF, Kashani HRK, Lorigooini Z, Filosa R, Khan FB, Masoudian N, Nabavi SM. Plant based food bioactives: A boon or bane for neurological disorders. Crit Rev Food Sci Nutr 2022; 64:3279-3325. [PMID: 36369694 DOI: 10.1080/10408398.2022.2131729] [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] [Indexed: 11/14/2022]
Abstract
Neurological disorders are the foremost occurring diseases across the globe resulting in progressive dysfunction, loss of neuronal structure ultimately cell death. Therefore, attention has been drawn toward the natural resources for the search of neuroprotective agents. Plant-based food bioactives have emerged as potential neuroprotective agents for the treatment of neurodegenerative disorders. This comprehensive review primarily focuses on various plant food bioactive, mechanisms, therapeutic targets, in vitro and in vivo studies in the treatment of neurological disorders to explore whether they are boon or bane for neurological disorders. In addition, the clinical perspective of plant food bioactives in neurological disorders are also highlighted. Scientific evidences point toward the enormous therapeutic efficacy of plant food bioactives in the prevention or treatment of neurological disorders. Nevertheless, identification of food bioactive components accountable for the neuroprotective effects, mechanism, clinical trials, and consolidation of information flow are warranted. Plant food bioactives primarily act by mediating through various pathways including oxidative stress, neuroinflammation, apoptosis, excitotoxicity, specific proteins, mitochondrial dysfunction, and reversing neurodegeneration and can be used for the prevention and therapy of neurodegenerative disorders. In conclusion, the plant based food bioactives are boon for neurological disorders.
Collapse
Affiliation(s)
- Neeraj Choudhary
- Department of Pharmacognosy, Adesh Institute of Pharmacy and Biomedical Sciences, Adesh University, Bathinda, Punjab, India
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Seyed Fazel Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
| | - Hamid Reza Khayat Kashani
- Department of Neurosurgery, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Rosanna Filosa
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Department of Science and Technology, University of Sannio, 82100, Benevento, Italy
| | - Farheen Badrealam Khan
- Department of Biology, College of Science, The United Arab Emirates University, Al Ain, 15551 United Arab Emirates
| | - Nooshin Masoudian
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
| | - Seyed Mohammad Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
| |
Collapse
|
12
|
Carmona Mata V, Goldberg J. Morin and isoquercitrin protect against ischemic neuronal injury by modulating signaling pathways and stimulating mitochondrial biogenesis. Nutr Neurosci 2022:1-11. [PMID: 35857717 DOI: 10.1080/1028415x.2022.2094855] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJETIVE The search for the etiology of Alzheimer's disease has revealed dysregulation of amyloid protein precursors, β-secretase, mitophagy, apoptosis, and Tau protein genes after ischemic brain injury. Due to this and the fact that some flavonoids have demonstrated anti-amyloidogenic effects on AD targets, we aimed to investigate whether they are effective against an ischemic neuronal injury not only by its antioxidant effects and clarify their mechanism.We simulated the energy depletion that characterizes ischemic processes using iodoacetic acid on HT22 cells. In vitro ischemic assays were also performed under OXPHOS inhibition using inhibitors of the different mitochondrial complexes and intracellular ATP, NADH and NADPH levels were determined. The signaling pathways of MAP kinase (MAPK) and of the PI3K/Akt mTOR were analyzed for its close association with post-ischemic survival. RESULTS Morin and isoquercitrin showed a significant neuroprotective effect against IAA toxicity, favored the activity of the mitochondrial complexes and prevented the decrease in ERK phosphorylation and activation of the stress proteins JNK and p38 caused by IAA treatment, as well as prevented satisfactorily mTOR and p70 dephosphorylation. They provide a considerable resistance to ischemic brain injury by modulating signaling pathways that stimulate mitochondrial biogenesis and promoting the activity of electron transport chain.
Collapse
Affiliation(s)
- Vanesa Carmona Mata
- Departamento de Farmacología, Farmacognosia y Botánica. Facultad de Farmacia, Universidad Complutense, Madrid, Spain.,Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Joshua Goldberg
- Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| |
Collapse
|
13
|
Hofmann J, Spatz P, Walther R, Gutmann M, Maurice T, Decker M. Synthesis and Biological Evaluation of Flavonoid‐Cinnamic Acid Amide Hybrids with Distinct Activity against Neurodegeneration in Vitro and in Vivo. Chemistry 2022; 28:e202200786. [PMID: 35621167 PMCID: PMC9400986 DOI: 10.1002/chem.202200786] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Indexed: 11/10/2022]
Abstract
Flavonoids are polyphenolic natural products and have shown significant potential as disease‐modifying agents against neurodegenerative disorders like Alzheimer's disease (AD), with activities even in vivo. Hybridization of the natural products taxifolin and silibinin with cinnamic acid led to an overadditive effect of these compounds in several phenotypic screening assays related to neurodegeneration and AD. Therefore, we have exchanged the flavonoid part of the hybrids with different flavonoids, which show higher efficacy than taxifolin or silibinin, to improve the activity of the respective hybrids. Chemical connection between the flavonoid and cinnamic acid was realized by an amide instead of a labile ester bond to improve stability towards hydrolysis. To investigate the influence of a double bond at the C‐ring of the flavonoid, the dehydro analogues of the respective hybrids were also synthesized. All compounds obtained show neuroprotection against oxytosis, ferroptosis and ATP‐depletion, respectively, in the murine hippocampal cell line HT22. Interestingly, the taxifolin and the quercetin derivatives are the most active compounds, whereby the quercetin derivate shows even more pronounced activity than the taxifolin one in all assays applied. As aimed for, no hydrolysis product was found in cellular uptake experiments after 4 h whereas different metabolites were detected. Furthermore, the quercetin‐cinnamic acid amide showed pronounced activity in an in vivo AD mouse model at a remarkably low dose of 0.3 mg/kg.
Collapse
Affiliation(s)
- Julian Hofmann
- Pharmaceutical and Medicinal Chemistry Institute of Pharmacy and Food Chemistry University of Würzburg Am Hubland 97074 Würzburg Germany
| | - Philipp Spatz
- Pharmaceutical and Medicinal Chemistry Institute of Pharmacy and Food Chemistry University of Würzburg Am Hubland 97074 Würzburg Germany
| | - Rasmus Walther
- Pharmaceutical and Medicinal Chemistry Institute of Pharmacy and Food Chemistry University of Würzburg Am Hubland 97074 Würzburg Germany
| | - Marcus Gutmann
- Drug Formulation and Delivery Institute of Pharmacy and Food Chemistry University of Würzburg Am Hubland 97074 Würzburg Germany
| | - Tangui Maurice
- MMDN University of Montpellier, EPHE, INSERM 34095 Montpellier France
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry Institute of Pharmacy and Food Chemistry University of Würzburg Am Hubland 97074 Würzburg Germany
| |
Collapse
|
14
|
Soriano-Castell D, Liang Z, Maher P, Currais A. Profiling the chemical nature of anti-oxytotic/ferroptotic compounds with phenotypic screening. Free Radic Biol Med 2021; 177:313-325. [PMID: 34748909 PMCID: PMC8639737 DOI: 10.1016/j.freeradbiomed.2021.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/26/2022]
Abstract
Because old age is the greatest risk factor for Alzheimer's disease (AD), it is critical to target the pathological events that link aging to AD in order to develop an efficient treatment that acts upon the primary causes of the disease. One such event might be the activation of oxytosis/ferroptosis, a unique cell death mechanism characterized by mitochondrial dysfunction and lethal lipid peroxidation. Here, a comprehensive library of >900 natural compounds was screened for protection against oxytosis/ferroptosis in nerve cells with the goal of better understanding the chemical nature of inhibitors of oxytosis/ferroptosis. Although the compounds tested spanned structurally diverse chemical classes from animal, microbial, plant and synthetic origins, a small set of very potent anti-oxytotic/ferroptotic compounds was identified that was highly enriched in plant quinones. The ability of these compounds to protect against oxytosis/ferroptosis strongly correlated with their ability to protect against in vitro ischemia and intracellular amyloid-beta toxicity in nerve cells, indicating that aspects of oxytosis/ferroptosis also underly other toxicities that are relevant to AD. Importantly, the anti-oxytotic/ferroptotic character of the quinone compounds relied on their capacity to target and directly prevent lipid peroxidation in a manner that required the reducing activity of cellular redox enzymes, such as NAD(P)H:quinone oxidoreductase 1 (NQO1) and ferroptosis suppressor protein 1 (FSP1). Because some of the compounds increased the production of total reactive oxygen species while decreasing lipid peroxidation, it appears that the pro-oxidant character of a compound can coexist with an inhibitory effect on lipid peroxidation and, consequently, still prevent oxytosis/ferroptosis. These findings have significant implications for the understanding of oxytosis/ferroptosis and open new approaches to the development of future neurotherapies.
Collapse
Affiliation(s)
- David Soriano-Castell
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd. La Jolla, CA, 92037, USA.
| | - Zhibin Liang
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd. La Jolla, CA, 92037, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd. La Jolla, CA, 92037, USA
| | - Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd. La Jolla, CA, 92037, USA.
| |
Collapse
|
15
|
Soriano-Castell D, Currais A, Maher P. Defining a pharmacological inhibitor fingerprint for oxytosis/ferroptosis. Free Radic Biol Med 2021; 171:219-231. [PMID: 34010663 PMCID: PMC8217321 DOI: 10.1016/j.freeradbiomed.2021.05.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 01/18/2023]
Abstract
Ferroptosis was first described in 2012 as an iron- and lipid peroxidation-dependent form of regulated cell death. Since its initial description, these two characteristics have informed numerous cell culture studies where inhibitors of lipid peroxidation and/or iron chelators have been shown to prevent cell death induced by a wide range of insults. However, it is not clear whether these two characteristics are sufficient to distinguish ferroptosis from other forms of regulated cell death. Thus, the primary goal of this study was to determine whether a unique combination of features could be identified that would provide an approach to more clearly separate ferroptosis from other forms of regulated cell death. To this end, multiple pharmacological inhibitors based on a variety of studies were tested. Many of these inhibitors were previously shown to protect cells from oxytosis, a regulated cell death pathway that mechanistically overlaps with ferroptosis and is induced by some of the same chemicals as ferroptosis. These inhibitors were not only tested against both known ferroptosis and oxytosis inducers but also a number of other insults that have been suggested to induce ferroptosis. The results show that a pharmacological fingerprint for ferroptosis can be established and used to categorize toxic insults into those that overlap with oxytosis/ferroptosis and those that do not.
Collapse
Affiliation(s)
- David Soriano-Castell
- Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Antonio Currais
- Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Pamela Maher
- Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA.
| |
Collapse
|
16
|
Cho YJ, Choi SH, Lee RM, Cho HS, Rhim H, Kim HC, Kim BJ, Kim JH, Nah SY. Protective Effects of Gintonin on Reactive Oxygen Species-Induced HT22 Cell Damages: Involvement of LPA1 Receptor-BDNF-AKT Signaling Pathway. Molecules 2021; 26:4138. [PMID: 34299412 PMCID: PMC8303475 DOI: 10.3390/molecules26144138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022] Open
Abstract
Gintonin is a kind of ginseng-derived glycolipoprotein that acts as an exogenous LPA receptor ligand. Gintonin has in vitro and in vivo neuroprotective effects; however, little is known about the cellular mechanisms underlying the neuroprotection. In the present study, we aimed to clarify how gintonin attenuates iodoacetic acid (IAA)-induced oxidative stress. The mouse hippocampal cell line HT22 was used. Gintonin treatment significantly attenuated IAA-induced reactive oxygen species (ROS) overproduction, ATP depletion, and cell death. However, treatment with Ki16425, an LPA1/3 receptor antagonist, suppressed the neuroprotective effects of gintonin. Gintonin elicited [Ca2⁺]i transients in HT22 cells. Gintonin-mediated [Ca2⁺]i transients through the LPA1 receptor-PLC-IP3 signaling pathway were coupled to increase both the expression and release of BDNF. The released BDNF activated the TrkB receptor. Induction of TrkB phosphorylation was further linked to Akt activation. Phosphorylated Akt reduced IAA-induced oxidative stress and increased cell survival. Our results indicate that gintonin attenuated IAA-induced oxidative stress in neuronal cells by activating the LPA1 receptor-BDNF-TrkB-Akt signaling pathway. One of the gintonin-mediated neuroprotective effects may be achieved via anti-oxidative stress in nervous systems.
Collapse
Affiliation(s)
- Yeon-Jin Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea; (Y.-J.C.); (S.-H.C.); (R.-M.L.); (H.-S.C.)
| | - Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea; (Y.-J.C.); (S.-H.C.); (R.-M.L.); (H.-S.C.)
| | - Ra-Mi Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea; (Y.-J.C.); (S.-H.C.); (R.-M.L.); (H.-S.C.)
| | - Han-Sung Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea; (Y.-J.C.); (S.-H.C.); (R.-M.L.); (H.-S.C.)
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul 02792, Korea;
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea;
| | - Byung-Joo Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan 50612, Korea;
| | - Jong-Hoon Kim
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Baekje-daero 567, Jeonju 28644, Korea;
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea; (Y.-J.C.); (S.-H.C.); (R.-M.L.); (H.-S.C.)
| |
Collapse
|
17
|
Abstract
Neurological disorders, including neurodegenerative diseases, have a significant negative impact on both patients and society at large. Since the prevalence of most of these disorders increases with age, the consequences for our aging population are only going to grow. It is now acknowledged that neurological disorders are multi-factorial involving disruptions in multiple cellular systems. While each disorder has specific initiating mechanisms and pathologies, certain common pathways appear to be involved in most, if not all, neurological disorders. Thus, it is becoming increasingly important to identify compounds that can modulate the multiple pathways that contribute to disease development or progression. One of these compounds is the flavonol fisetin. Fisetin has now been shown in preclinical models to be effective at preventing the development and/or progression of multiple neurological disorders including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, stroke (both ischemic and hemorrhagic) and traumatic brain injury as well as to reduce age-associated changes in the brain. These beneficial effects stem from its actions on multiple pathways associated with the different neurological disorders. These actions include its well characterized anti-inflammatory and anti-oxidant effects as well as more recently described effects on the regulated cell death oxytosis/ferroptosis pathway, the gut microbiome and its senolytic activity. Therefore, the growing body of pre-clinical data, along with fisetin’s ability to modulate a large number of pathways associated with brain dysfunction, strongly suggest that it would be worthwhile to pursue its therapeutic effects in humans.
Collapse
Affiliation(s)
- Pamela Maher
- Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA
| |
Collapse
|
18
|
Maher P. Investigations into the Role of Metabolism in the Inflammatory Response of BV2 Microglial Cells. Antioxidants (Basel) 2021; 10:109. [PMID: 33466581 PMCID: PMC7828726 DOI: 10.3390/antiox10010109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 01/16/2023] Open
Abstract
Although the hallmarks of Alzheimer's disease (AD) are amyloid beta plaques and neurofibrillary tangles, there is growing evidence that neuroinflammation, mitochondrial dysfunction and oxidative stress play important roles in disease development and progression. A major risk factor for the development of AD is diabetes, which is also characterized by oxidative stress and mitochondrial dysfunction along with chronic, low-grade inflammation. Increasing evidence indicates that in immune cells, the induction of a pro-inflammatory phenotype is associated with a shift from oxidative phosphorylation (OXPHOS) to glycolysis. However, whether hyperglycemia also contributes to this shift is not clear. Several different approaches including culturing BV2 microglial cells in different carbon sources, using enzyme inhibitors and knocking down key pathway elements were used in conjunction with bacterial lipopolysaccharide (LPS) activation to address this question. The results indicate that while high glucose favors NO production, pro-inflammatory cytokine production is highest in the presence of carbon sources that drive OXPHOS. In addition, among the carbon sources that drive OXPHOS, glutamine is a very potent inducer of IL6 production. This effect is dampened in the presence of glucose. Together, these results may provide new prospects for the therapeutic manipulation of neuroinflammation in the context of diabetes and AD.
Collapse
Affiliation(s)
- Pamela Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
19
|
Parrella E, Gussago C, Porrini V, Benarese M, Pizzi M. From Preclinical Stroke Models to Humans: Polyphenols in the Prevention and Treatment of Stroke. Nutrients 2020; 13:nu13010085. [PMID: 33383852 PMCID: PMC7823436 DOI: 10.3390/nu13010085] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Polyphenols are an important family of molecules of vegetal origin present in many medicinal and edible plants, which represent important alimentary sources in the human diet. Polyphenols are known for their beneficial health effects and have been investigated for their potential protective role against various pathologies, including cancer, brain dysfunctions, cardiovascular diseases and stroke. The prevention of stroke promoted by polyphenols relies mainly on their effect on cardio- and cerebrovascular systems. However, a growing body of evidence from preclinical models of stroke points out a neuroprotective role of these molecules. Notably, in many preclinical studies, the polyphenolic compounds were effective also when administered after the stroke onset, suggesting their possible use in promoting recovery of patients suffering from stroke. Here, we review the effects of the major polyphenols in cellular and in vivo models of both ischemic and hemorrhagic stroke in immature and adult brains. The results from human studies are also reported.
Collapse
|
20
|
Maher P, Currais A, Schubert D. Using the Oxytosis/Ferroptosis Pathway to Understand and Treat Age-Associated Neurodegenerative Diseases. Cell Chem Biol 2020; 27:1456-1471. [PMID: 33176157 PMCID: PMC7749085 DOI: 10.1016/j.chembiol.2020.10.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/31/2020] [Accepted: 10/20/2020] [Indexed: 12/18/2022]
Abstract
Oxytosis was first described over 30 years ago in nerve cells as a non-excitotoxic pathway for glutamate-induced cell death. The key steps of oxytosis, including glutathione depletion, lipoxygenase activation, reactive oxygen species accumulation, and calcium influx, were identified using a combination of chemical and genetic tools. A pathway with the same characteristics as oxytosis was identified in transformed fibroblasts in 2012 and named ferroptosis. Importantly, the pathophysiological changes seen in oxytosis and ferroptosis are also observed in multiple neurodegenerative diseases as well as in the aging brain. This led to the hypothesis that this pathway could be used as a screening tool to identify novel drug candidates for the treatment of multiple age-associated neurological disorders, including Alzheimer's disease (AD). Using this approach, we have identified several AD drug candidates, one of which is now in clinical trials, as well as new target pathways for AD.
Collapse
Affiliation(s)
- Pamela Maher
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Antonio Currais
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - David Schubert
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
21
|
Yu Y, Wang F, Wang J, Zhang D, Zhao X. Ketogenic diet attenuates aging-associated myocardial remodeling and dysfunction in mice. Exp Gerontol 2020; 140:111058. [DOI: 10.1016/j.exger.2020.111058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 01/06/2023]
|
22
|
Maher PA. Using Plants as a Source of Potential Therapeutics for the Treatment of Alzheimer's Disease. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:365-373. [PMID: 32607095 PMCID: PMC7309672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia with the numbers expected to increase dramatically as our society ages. There are no treatments to cure, prevent, or slow down the progression of the disease. Age is the single greatest risk factor for AD. However, to date, AD drug discovery efforts have generally not taken this fact into consideration. Multiple changes associated with brain aging, including neuroinflammation and oxidative stress, are important contributors to disease development and progression. Thus, due to the multifactorial nature of AD, the one target strategy to fight the disease needs to be replaced by a more general approach using pleiotropic compounds to deal with the complexity of the disease. In this perspectives piece, our alternative approach to AD drug development based on the biology of aging is described. Starting with plants or plant-derived natural products, we have used a battery of cell-based screening assays that reflect multiple, age-associated toxicity pathways to identify compounds that can target the aspects of aging that contribute to AD pathology. We have found that this combination of assays provides a replicable, cost- and time-effective screening approach that has to date yielded one compound in clinical trials for AD (NCT03838185) and several others that show significant promise.
Collapse
Affiliation(s)
- Pamela A. Maher
- To whom all correspondence should be addressed: Pamela A. Maher, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037; Tel: 858-453-4100 x1932;
| |
Collapse
|
23
|
Wang L, Cao D, Wu H, Jia H, Yang C, Zhang L. Fisetin Prolongs Therapy Window of Brain Ischemic Stroke Using Tissue Plasminogen Activator: A Double-Blind Randomized Placebo-Controlled Clinical Trial. Clin Appl Thromb Hemost 2020; 25:1076029619871359. [PMID: 31434498 PMCID: PMC6829632 DOI: 10.1177/1076029619871359] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Recombinant tissue plasminogen activator (rt-PA) can be utilized to treat ischemic stroke
with safety and effectiveness but limited by a narrow therapeutic window. In the present
clinical trial among patients with stroke, we sought to evaluate the potential of fisetin
to extend the therapeutic window of rt-PA treatment. Patients with stroke were divided
based on their onset-to-treatment time (OTT) and then randomly assigned to receive the
rt-PA treatment combined with fisetin or placebo. Primary outcome was evaluated using the
National Institutes of Health Stroke scale (NIHSS), and secondary outcome was assessed by
serum levels of matrix metalloproteinase (MMP) 2, MMP 9, and C-reactive protein (CRP).
Fisetin dramatically improved the treatment outcomes of the patients with stroke in the
delayed OTT strata, as revealed by lower NIHSS scores. The beneficial effect of fisetin
was likely attributable to reduced levels of MMP-2, MMP-9, and CRP in the serum, as
evidenced by strong linear correlations between serum levels of such markers with the
NIHSS scores in all enrolled patients. Fisetin may possess the potential to supplement
traditional rt-PA treatments among patients with stroke, particularly for those with
delayed OTT, and thereby extend the otherwise narrow therapeutic window and improve the
treatment outcomes.
Collapse
Affiliation(s)
- Limin Wang
- 1 Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Di Cao
- 1 Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Huijun Wu
- 1 Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Hongning Jia
- 1 Cangzhou Central Hospital, Cangzhou, Hebei, China
| | | | - Lihua Zhang
- 1 Cangzhou Central Hospital, Cangzhou, Hebei, China
| |
Collapse
|
24
|
7-O-Esters of taxifolin with pronounced and overadditive effects in neuroprotection, anti-neuroinflammation, and amelioration of short-term memory impairment in vivo. Redox Biol 2019; 29:101378. [PMID: 31926632 PMCID: PMC6928325 DOI: 10.1016/j.redox.2019.101378] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is a multifactorial disease and the most common form of dementia. There are no treatments to cure, prevent or slow down the progression of the disease. Natural products hold considerable interest for the development of preventive neuroprotectants to treat neurodegenerative disorders like AD, due to their low toxicity and general beneficial effects on human health with their anti-inflammatory and antioxidant features. In this work we describe regioselective synthesis of 7-O-ester hybrids of the flavonoid taxifolin with the phenolic acids cinnamic and ferulic acid, namely 7-O-cinnamoyltaxifolin and 7-O-feruloyltaxifolin. The compounds show pronounced overadditive neuroprotective effects against oxytosis, ferroptosis and ATP depletion in the murine hippocampal neuron HT22 cell model. Furthermore, 7-O-cinnamoyltaxifolin and 7-O-feruloyltaxifolin reduced LPS-induced neuroinflammation in BV-2 microglia cells as assessed by effects on the levels of NO, IL6 and TNFα. In all in vitro assays the 7-O-esters of taxifolin and ferulic or cinnamic acid showed strong overadditive activity, significantly exceeding the effects of the individual components and the equimolar mixtures thereof, which were almost inactive in all of the assays at the tested concentrations. In vivo studies confirmed this overadditive effect. Treatment of an AD mouse model based on the injection of oligomerized Aβ25-35 peptide into the brain to cause neurotoxicity and subsequently memory deficits with 7-O-cinnamoyltaxifolin or 7-O-feruloyltaxifolin resulted in improved performance in an assay for short-term memory as compared to vehicle and mice treated with the respective equimolar mixtures. These results highlight the benefits of natural product hybrids as a novel compound class with potential use for drug discovery in neurodegenerative diseases due to their pharmacological profile that is distinct from the individual natural components.
Collapse
|
25
|
Schubert D, Kepchia D, Liang Z, Dargusch R, Goldberg J, Maher P. Efficacy of Cannabinoids in a Pre-Clinical Drug-Screening Platform for Alzheimer's Disease. Mol Neurobiol 2019; 56:7719-7730. [PMID: 31104297 PMCID: PMC6815693 DOI: 10.1007/s12035-019-1637-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022]
Abstract
Finding a therapy for Alzheimer's disease (AD) is perhaps the greatest challenge for modern medicine. The chemical scaffolds of many drugs in the clinic today are based upon natural products from plants, yet Cannabis has not been extensively examined as a source of potential AD drug candidates. Here, we determine if a number of non-psychoactive cannabinoids are neuroprotective in a novel pre-clinical AD and neurodegeneration drug-screening platform that is based upon toxicities associated with the aging brain. This drug discovery paradigm has yielded several compounds in or approaching clinical trials for AD. Eleven cannabinoids were assayed for neuroprotection in assays that recapitulate proteotoxicity, loss of trophic support, oxidative stress, energy loss, and inflammation. These compounds were also assayed for their ability to remove intraneuronal amyloid and subjected to a structure-activity relationship analysis. Pairwise combinations were assayed for their ability to synergize to produce neuroprotective effects that were greater than additive. Nine of the 11 cannabinoids have the ability to protect cells in four distinct phenotypic neurodegeneration screening assays, including those using neurons that lack CB1 and CB2 receptors. They are able to remove intraneuronal Aβ, reduce oxidative damage, and protect from the loss of energy or trophic support. Structure-activity relationship (SAR) data show that functional antioxidant groups such as aromatic hydroxyls are necessary but not sufficient for neuroprotection. Therefore, there is a need to focus upon CB1 agonists that have these functionalities if neuroprotection is the goal. Pairwise combinations of THC and CBN lead to a synergistic neuroprotective interaction. Together, these results significantly extend the published data by showing that non-psychoactive cannabinoids are potential lead drug candidates for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- David Schubert
- Cellular Neurobiological Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA
| | - Devin Kepchia
- Cellular Neurobiological Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA
| | - Zhibin Liang
- Cellular Neurobiological Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA
| | - Richard Dargusch
- Cellular Neurobiological Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA
| | | | - Pamela Maher
- Cellular Neurobiological Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA.
| |
Collapse
|
26
|
Linalool attenuates oxidative stress and mitochondrial dysfunction mediated by glutamate and NMDA toxicity. Biomed Pharmacother 2019; 118:109295. [DOI: 10.1016/j.biopha.2019.109295] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/17/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
|
27
|
Elkholy R, Balaha M, El-Anwar N, Kandeel S, Hedya S, Abd-El Rahman MN. Fisetin and telmisartan each alone or in low-dose combination alleviate OVA-induced food allergy in mice. Pharmacol Rep 2019; 71:330-337. [PMID: 30826574 DOI: 10.1016/j.pharep.2018.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/17/2018] [Accepted: 12/20/2018] [Indexed: 01/23/2023]
Abstract
BACKGROUND Food allergy (FA) is a worldwide health problem, affecting nearly 10% of all populations, with no prophylactic options or regulatory treatment available until now. Fisetin, a biologically active flavonoid, and telmisartan, the highly selective competitive AT1 receptor antagonist, recently exhibited potent anti-inflammatory and immunomodulatory activities. In the present study, we have evaluated the possible anti-inflammatory and immunomodulatory activities of fisetin and telmisartan each alone or in low-dose combination in a mouse model of FA. METHODS For induction of FA, eight-week-old BALB/c mice, sensitized by two ip injection of 50 μg ovalbumin (OVA) and 1 mg alum at day 0 and 7. Then, each mouse challenged with 10 mg OVA at days 14, 16, 18, and 21. On the 28th day, the fifth challenge carried out by oral administration of 50 mg OVA. Either fisetin (1 or 3 mg/kg/d), telmisartan (1 or 3 mg/kg/d) or a combination of fisetin 1 mg/kg/d and telmisartan 1 mg/kg/d received orally from the 13th day till 28th day. In challenge days, the treatments received one-hour before the challenge. RESULTS Our data showed that fisetin and telmisartan each alone or in low-dose combination attenuated the anaphylactic manifestation, decreased blood eosinophilic count, serum OVA-specific IgE, and IL-4 levels, the intestinal total and degranulated mast cells count, and CD4+ immunohistochemical expression. Furthermore, they enhanced the serum IFN-γ level and abrogated the intestinal histopathological changes induced by OVA in mice. CONCLUSION Either fisetin, telmisartan or their low-dose combination could be promising in the management of FA.
Collapse
Affiliation(s)
- Reem Elkholy
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohamed Balaha
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Saudi Arabia; Pharmacology Department, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | - Noha El-Anwar
- Pathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Samah Kandeel
- Histology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Sabiha Hedya
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohamed-Nabih Abd-El Rahman
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta, Egypt; Pharmacology Department, Faculty of Medicine, Bisha University, Saudi Arabia
| |
Collapse
|
28
|
Han Y, Qu P, Zhang K, Bi Y, Zhou L, Xie D, Song H, Dong J, Qi J. Storage solution containing hydrogen improves the preservation effect of osteochondral allograft. Cell Tissue Bank 2019; 20:201-208. [DOI: 10.1007/s10561-019-09758-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 02/21/2019] [Indexed: 12/26/2022]
|
29
|
Fokialakis N, Alexi X, Aligiannis N, Boulaka A, Meligova AK, Lambrinidis G, Kalpoutzakis E, Pratsinis H, Cheilari A, Mitsiou DJ, Mitakou S, Alexis MN. Biological evaluation of isoflavonoids from Genista halacsyi using estrogen-target cells: Activities of glucosides compared to aglycones. PLoS One 2019; 14:e0210247. [PMID: 30620769 PMCID: PMC6324813 DOI: 10.1371/journal.pone.0210247] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/19/2018] [Indexed: 12/21/2022] Open
Abstract
The purpose of this study was to evaluate the response of estrogen target cells to a series of isoflavone glucosides and aglycones from Genista halacsyi Heldr. The methanolic extract of aerial parts of this plant was processed using fast centrifugal partition chromatography, resulting in isolation of four archetypal isoflavones (genistein, daidzein, isoprunetin, 8-C-β-D-glucopyranosyl-genistein) and ten derivatives thereof. 7-O-β-D-glucopyranosyl-genistein and 7,4΄-di-O-β-D-glucopyranosyl-genistein were among the most abundant constituents of the isolate. All fourteen, except genistein, displayed low binding affinity for estrogen receptors (ER). Models of binding to ERα could account for the low binding affinity of monoglucosides. Genistein and its glucosides displayed full efficacy in inducing alkaline phosphatase (AlkP) in Ishikawa cells, proliferation of MCF-7 cells and ER-dependent gene expression in reporter cells at low concentrations (around 0.3 μM). ICI182,780 fully antagonized these effects. The AlkP-inducing efficacy of the fourteen isoflavonoids was more strongly correlated with their transcriptional efficacy through ERα. O-monoglucosides displayed higher area under the dose-response curve (AUC) of AlkP response relative to the AUC of ERα-transcriptional response compared to the respective aglycones. In addition, 7-O-β-D-glucopyranosyl-genistein and 7,4΄-di-O-β-D-glucopyranosyl-genistein displayed estradiol-like efficacy in promoting differentiation of MC3T3-E1 cells to osteoblasts, while genistein was not convincingly effective in this respect. Moreover, 7,4΄-di-O-β-D-glucopyranosyl-genistein suppressed lipopolysaccharide-induced tumor necrosis factor mRNA expression in RAW 264.7 cells, while 7-O-β-D-glucopyranosyl-genistein was not convincingly effective and genistein was ineffective. However, genistein and its O-glucosides were ineffective in inhibiting differentiation of RAW 264.7 cells to osteoclasts and in protecting glutamate-challenged HT22 hippocampal neurons from oxidative stress-induced cell death. These findings suggest that 7-O-β-D-glucopyranosyl-genistein and 7,4΄-di-O-β-D-glucopyranosyl-genistein display higher estrogen-like and/or anti-inflammatory activity compared to the aglycone. The possibility of using preparations rich in O-β-D-glucopyranosides of genistein to substitute for low-dose estrogen in formulations for menopausal symptoms is discussed.
Collapse
Affiliation(s)
- Nikolas Fokialakis
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
- * E-mail: (MNA); (NF)
| | - Xanthippi Alexi
- Molecular Endocrinology Program, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Nektarios Aligiannis
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Athina Boulaka
- Molecular Endocrinology Program, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Aggeliki K. Meligova
- Molecular Endocrinology Program, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - George Lambrinidis
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleftherios Kalpoutzakis
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Harris Pratsinis
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences & Applications, NCSR "Demokritos", Athens, Greece
| | - Antigoni Cheilari
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitra J. Mitsiou
- Molecular Endocrinology Program, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Sofia Mitakou
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael N. Alexis
- Molecular Endocrinology Program, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
- * E-mail: (MNA); (NF)
| |
Collapse
|
30
|
Old age-associated phenotypic screening for Alzheimer's disease drug candidates identifies sterubin as a potent neuroprotective compound from Yerba santa. Redox Biol 2018; 21:101089. [PMID: 30594901 PMCID: PMC6309122 DOI: 10.1016/j.redox.2018.101089] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the most frequent age-associated dementia with no treatments that can prevent or slow its progression. Since age is by far the major risk factor for AD, there is a strong rationale for an alternative approach to drug discovery based upon the biology of aging. Phenotypic screening assays that reflect multiple, age-associated neurotoxicity pathways rather than single molecular targets can identify compounds that have therapeutic efficacy by targeting aspects of aging that contribute to AD pathology. And, while the suitability of any single assay can be questioned, a combination of assays can make reliable predictions about the neuroprotective effects of compounds in vivo. Therefore, our lab has developed a combination of phenotypic screening assays that are ideally suited not only to identify novel neuroprotective compounds for the treatment of AD but also their target pathways, thereby potentially providing new therapeutic targets for disease treatment. Using these assays, we screened a large library of extracts from plants with identified pharmacological uses. Analysis of one of these extracts from the plant Yerba santa (Eriodictyon californicum) identified the flavanone sterubin as the active component and further studies showed it to be a potent neuroprotective and anti-inflammatory compound. Phenotypic screening of a curated library of plant extracts identifies Yerba santa. The flavonoid sterubin is the main active component of Yerba santa. Sterubin is very neuroprotective against multiple toxicities of the aging brain. Sterubin has potent anti-inflammatory activity that is dependent on Nrf2 induction. Sterubin is also an iron chelator which could enhance its neuroprotective activity.
Collapse
|
31
|
Balasubramanian PK, Kim J, Son K, Durai P, Kim Y. 3,6-Dihydroxyflavone: A Potent Inhibitor with Anti-Inflammatory Activity Targeting Toll-like Receptor 2. B KOREAN CHEM SOC 2018. [DOI: 10.1002/bkcs.11644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
| | - Jieun Kim
- Department of Bioscience and Biotechnology; Konkuk University; Seoul 05029 South Korea
| | - Kkabi Son
- Department of Bioscience and Biotechnology; Konkuk University; Seoul 05029 South Korea
| | | | - Yangmee Kim
- Department of Bioscience and Biotechnology; Konkuk University; Seoul 05029 South Korea
| |
Collapse
|
32
|
Goldberg J, Currais A, Prior M, Fischer W, Chiruta C, Ratliff E, Daugherty D, Dargusch R, Finley K, Esparza‐Moltó PB, Cuezva JM, Maher P, Petrascheck M, Schubert D. The mitochondrial ATP synthase is a shared drug target for aging and dementia. Aging Cell 2018; 17:e12715. [PMID: 29316249 PMCID: PMC5847861 DOI: 10.1111/acel.12715] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2017] [Indexed: 12/31/2022] Open
Abstract
Aging is a major driving force underlying dementia, such as that caused by Alzheimer's disease (AD). While the idea of targeting aging as a therapeutic strategy is not new, it remains unclear how closely aging and age-associated diseases are coupled at the molecular level. Here, we discover a novel molecular link between aging and dementia through the identification of the molecular target for the AD drug candidate J147. J147 was developed using a series of phenotypic screening assays mimicking disease toxicities associated with the aging brain. We have previously demonstrated the therapeutic efficacy of J147 in several mouse models of AD. Here, we identify the mitochondrial α-F1 -ATP synthase (ATP5A) as a target for J147. By targeting ATP synthase, J147 causes an increase in intracellular calcium leading to sustained calcium/calmodulin-dependent protein kinase kinase β (CAMKK2)-dependent activation of the AMPK/mTOR pathway, a canonical longevity mechanism. Accordingly, modulation of mitochondrial processes by J147 prevents age-associated drift of the hippocampal transcriptome and plasma metabolome in mice and extends lifespan in drosophila. Our results link aging and age-associated dementia through ATP synthase, a molecular drug target that can potentially be exploited for the suppression of both. These findings demonstrate that novel screens for new AD drug candidates identify compounds that act on established aging pathways, suggesting an unexpectedly close molecular relationship between the two.
Collapse
Affiliation(s)
- Joshua Goldberg
- Cellular NeurobiologyThe Salk Institute for Biological StudiesLa JollaCAUSA
| | - Antonio Currais
- Cellular NeurobiologyThe Salk Institute for Biological StudiesLa JollaCAUSA
| | - Marguerite Prior
- Cellular NeurobiologyThe Salk Institute for Biological StudiesLa JollaCAUSA
| | - Wolfgang Fischer
- Cellular NeurobiologyThe Salk Institute for Biological StudiesLa JollaCAUSA
| | | | - Eric Ratliff
- Donald P. Shiley BioScience CenterSan Diego State UniversitySan DiegoCAUSA
| | - Daniel Daugherty
- Cellular NeurobiologyThe Salk Institute for Biological StudiesLa JollaCAUSA
| | - Richard Dargusch
- Cellular NeurobiologyThe Salk Institute for Biological StudiesLa JollaCAUSA
| | - Kim Finley
- Donald P. Shiley BioScience CenterSan Diego State UniversitySan DiegoCAUSA
| | | | - José M. Cuezva
- Centro de Biología MolecularCIBERER, Universidad Autónoma de MadridMadridSpain
| | - Pamela Maher
- Cellular NeurobiologyThe Salk Institute for Biological StudiesLa JollaCAUSA
| | | | - David Schubert
- Cellular NeurobiologyThe Salk Institute for Biological StudiesLa JollaCAUSA
| |
Collapse
|
33
|
Currais A, Farrokhi C, Dargusch R, Armando A, Quehenberger O, Schubert D, Maher P. Fisetin Reduces the Impact of Aging on Behavior and Physiology in the Rapidly Aging SAMP8 Mouse. J Gerontol A Biol Sci Med Sci 2018; 73:299-307. [PMID: 28575152 PMCID: PMC5861950 DOI: 10.1093/gerona/glx104] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 05/26/2017] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease (AD) is rarely addressed in the context of aging even though there is an overlap in pathology. We previously used a phenotypic screening platform based on old age-associated brain toxicities to identify the flavonol fisetin as a potential therapeutic for AD and other age-related neurodegenerative diseases. Based on earlier results with fisetin in transgenic AD mice, we hypothesized that fisetin would be effective against brain aging and cognitive dysfunction in rapidly aging senescence-accelerated prone 8 (SAMP8) mice, a model for sporadic AD and dementia. An integrative approach was used to correlate protein expression and metabolite levels in the brain with cognition. It was found that fisetin reduced cognitive deficits in old SAMP8 mice while restoring multiple markers associated with impaired synaptic function, stress, and inflammation. These results provide further evidence for the potential benefits of fisetin for the treatment of age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Antonio Currais
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, California
| | - Catherine Farrokhi
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, California
| | - Richard Dargusch
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, California
| | - Aaron Armando
- Department of Medicine, University of California San Diego, La Jolla
| | | | - David Schubert
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, California
| | - Pamela Maher
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, California
| |
Collapse
|
34
|
Maher P. Protective effects of fisetin and other berry flavonoids in Parkinson's disease. Food Funct 2018; 8:3033-3042. [PMID: 28714503 DOI: 10.1039/c7fo00809k] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is an age-associated degenerative disease of the midbrain that results from the loss of dopaminergic neurons in the substantia nigra. It initially presents as a movement disorder with cognitive and other behavioral problems appearing later in the progression of the disease. Current therapies for PD only delay the onset or reduce the motor symptoms. There are no treatments to stop the nerve cell death or to cure the disease. It is becoming increasingly clear that neurological diseases such as PD are multi-factorial involving disruptions in multiple cellular systems. Thus, it is unlikely that modulating only a single factor will be effective at either preventing disease development or slowing disease progression. A better approach is to identify small molecules that have multiple biological activities relevant to the maintenance of brain function. Flavonoids are polyphenolic compounds that are widely distributed in fruits and vegetables and therefore regularly consumed in the human diet. While flavonoids were historically characterized on the basis of their antioxidant and free radical scavenging effects, more recent studies have shown that flavonoids have a wide range of activities that could make them particularly effective as agents for the treatment of PD. In this article, the multiple physiological benefits of flavonoids in the context of PD are first reviewed. Then, the evidence for the beneficial effects of the flavonol fisetin in models of PD are discussed. These results, coupled with the known actions of fisetin, suggest that it could reduce the impact of PD on brain function.
Collapse
Affiliation(s)
- Pamela Maher
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
35
|
Abstract
Hydrogen is the most abundant chemical element in the universe, and has been used as an inert gas for a long time. More recent studies have shown that molecular hydrogen as a kind of antioxidant, anti-inflammatory, anti-apoptosis, gene expression and signal modulation molecule, can be used for the treatment of many diseases. This review mainly focuses on the research progresses of hydrogen in various medical fields and the possible action mechanisms.
Collapse
Affiliation(s)
- Hong-Mei Li
- Department of Cardiothoracic Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Li Shen
- Department of Cardiothoracic Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun-Wen Ge
- Department of Cardiothoracic Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ru-Fang Zhang
- Department of Cardiothoracic Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
36
|
Antioxidant properties of the flavonoid fisetin: An updated review of in vivo and in vitro studies. Trends Food Sci Technol 2017. [DOI: 10.1016/j.tifs.2017.10.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
37
|
Daugherty D, Goldberg J, Fischer W, Dargusch R, Maher P, Schubert D. A novel Alzheimer's disease drug candidate targeting inflammation and fatty acid metabolism. ALZHEIMERS RESEARCH & THERAPY 2017; 9:50. [PMID: 28709449 PMCID: PMC5513091 DOI: 10.1186/s13195-017-0277-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/16/2017] [Indexed: 12/26/2022]
Abstract
Background CAD-31 is an Alzheimer’s disease (AD) drug candidate that was selected on the basis of its ability to stimulate the replication of human embryonic stem cell-derived neural precursor cells as well as in APPswe/PS1ΔE9 AD mice. To move CAD-31 toward the clinic, experiments were undertaken to determine its neuroprotective and pharmacological properties, as well as to assay its therapeutic efficacy in a rigorous mouse model of AD. Results CAD-31 has potent neuroprotective properties in six distinct nerve cell assays that mimic toxicities observed in the old brain. Pharmacological and preliminary toxicological studies show that CAD-31 is brain-penetrant and likely safe. When fed to old, symptomatic APPswe/PS1ΔE9 AD mice starting at 10 months of age for 3 additional months in a therapeutic model of the disease, there was a reduction in the memory deficit and brain inflammation, as well as an increase in the expression of synaptic proteins. Small-molecule metabolic data from the brain and plasma showed that the major effect of CAD-31 is centered on fatty acid metabolism and inflammation. Pathway analysis of gene expression data showed that CAD-31 had major effects on synapse formation and AD energy metabolic pathways. Conclusions All of the multiple physiological effects of CAD-31 were favorable in the context of preventing some of the toxic events in old age-associated neurodegenerative diseases. Electronic supplementary material The online version of this article (doi:10.1186/s13195-017-0277-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel Daugherty
- Cellular Neuroendocrinology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA
| | - Joshua Goldberg
- Cellular Neuroendocrinology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA
| | - Wolfgang Fischer
- Cellular Neuroendocrinology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA
| | - Richard Dargusch
- Cellular Neuroendocrinology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA
| | - Pamela Maher
- Cellular Neuroendocrinology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA
| | - David Schubert
- Cellular Neuroendocrinology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037-1002, USA.
| |
Collapse
|
38
|
Wang H, Xu YS, Wang ML, Cheng C, Bian R, Yuan H, Wang Y, Guo T, Zhu LL, Zhou H. Protective effect of naringin against the LPS-induced apoptosis of PC12 cells: Implications for the treatment of neurodegenerative disorders. Int J Mol Med 2017; 39:819-830. [PMID: 28260042 PMCID: PMC5360435 DOI: 10.3892/ijmm.2017.2904] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/10/2017] [Indexed: 01/01/2023] Open
Abstract
Several studies have demonstrated that increased apoptosis plays an essential role in neurodegenerative disorders. It has been demonstrated that lipopolysaccharide (LPS) induces apoptosis largely through the production of intracellular reactive oxygen species (ROS) and inflammatory mediators. In this study, we investigated the potential protective mechanisms of naringin (Nar), a pummelo peel extract, on LPS-induced PC12 cell apoptosis. Nar pre-conditioning prior to stimulation with LPS for 18 h was a prerequisite for evaluating PC12 cell viability and the protective mechanisms of Nar. Nar significantly improved cell survival in a time- and concentration-dependent manner. On the one hand, Nar downregulated cytochrome P450 2E1 (CYP2E1), inhibited the release of ROS, mitigated the stimulation of oxidative stress, and rectified the antioxidant protein contents of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), superoxide dismutase (SOD)2 and glutathione synthetase (GSS). On the other hand, Nar down-regulated inflammatory gene and protein expression, including interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, HMGB1, high mobility group box 1 protein (HMGB1), cyclo-oxygenase-2 (COX-2), the Toll-like receptor 4 (TLR4)-myeloid differentiation factor 88 (MyD88)-TNF receptor-associated factor 6 (TRAF6) path way and downstream mitogen activated protein kinase (MAPK) phosphorylation, activator protein transcription factor-1 (AP-1) and nuclear factor (NF)-κB. Moroever, Nar markedly attenuated the cytochrome c shift from the mitochondria to the cytosol and regulated caspase-3-related protein expression. To the best of our knowledge, this is the first study to report the antioxidant, anti-inflammatory and anti-apoptotic effects of Nar in neuronal-like PC12 cells. These results suggest that Nar can be utilized as a potential drug for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Hui Wang
- Department of Neurosurgery, Traffic Hospital of Shandong Province, Jinan, Shandong 250031, P.R. China
| | - You Song Xu
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Miao Lin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Chao Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Rui Bian
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Hao Yuan
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yi Wang
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Ting Guo
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Lin Lin Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Hang Zhou
- Department of Neurosurgery, The 2nd Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| |
Collapse
|
39
|
Yu S, Zhao C, Che N, Jing L, Ge R. Hydrogen-rich saline attenuates eosinophil activation in a guinea pig model of allergic rhinitis via reducing oxidative stress. JOURNAL OF INFLAMMATION-LONDON 2017; 14:1. [PMID: 28100959 PMCID: PMC5237150 DOI: 10.1186/s12950-016-0148-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/09/2016] [Indexed: 12/20/2022]
Abstract
Background It is well considered that reactive oxygen species (ROS) plays a prominent causative role in the development of allergic rhinitis (AR), and eosinophils cells as important allergic inflammatory cells contribute to elevating oxidative stress. Hydrogen, emerging as a novel antioxidant, has been proven effective in selectively reducing ROS in animals models of oxidative damage. We herein aim to verify protective effects of hydrogen on eosinophils cells in guinea pigs models of AR. Methods Thirty two guinea pigs were random divided into four groups, and AR model was established through ovalbumin sensitization. The guinea pigs were injected with hydrogen-rich saline (Normal-HRS and AR-HRS group) or normal saline (control and AR group). The frequencies of sneezing and scratching were recorded. The IgE level, blood eosinophil count and eosinophil cationic protein (ECP) level in serum were measured. The serum malondialdehyde (MDA) and superoxide dismutase (SOD) assays were also measured to evaluate oxidative stress. The expression levels of eotaxin mRNA and protein in the nasal mucosa were also determined by real-time RT-PCR, Western blot and immunofluorescence. Results HRS reduced the ROS and MDA levels and increased SOD level in guinea pigs of AR-HRS group accompanied with decreased frequency of sneezing and scratches. Meanwhile, there was a decline of the number of eosinophils cells in blood and of thelevel of ECP in serum in the AR-HRS group. HRS also significantly decreased the expression of eotaxin in nasal mucosa. Conclusion HRS may play a protective role in attenuating allergic inflammation, and suppressing the increase and activation of eosinophils in AR possibly through antioxidation effect of hydrogen.
Collapse
Affiliation(s)
- Shaoqing Yu
- Department of Otolaryngology, Tongji Hospital, Tongji University, 389 Xincun road, Putuo District, Shanghai, 200065 China
| | - Chuanliang Zhao
- Department of Otolaryngology, Tongji Hospital, Tongji University, 389 Xincun road, Putuo District, Shanghai, 200065 China
| | - Na Che
- Department of Otolaryngology, Tongji Hospital, Tongji University, 389 Xincun road, Putuo District, Shanghai, 200065 China
| | - Lin Jing
- Department of Otolaryngology, Tongji Hospital, Tongji University, 389 Xincun road, Putuo District, Shanghai, 200065 China
| | - Rongming Ge
- Department of Otolaryngology, Tongji Hospital, Tongji University, 389 Xincun road, Putuo District, Shanghai, 200065 China
| |
Collapse
|
40
|
Pazoki-Toroudi H, Amani H, Ajami M, Nabavi SF, Braidy N, Kasi PD, Nabavi SM. Targeting mTOR signaling by polyphenols: A new therapeutic target for ageing. Ageing Res Rev 2016; 31:55-66. [PMID: 27453478 DOI: 10.1016/j.arr.2016.07.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 06/19/2016] [Accepted: 07/15/2016] [Indexed: 12/19/2022]
Abstract
Current ageing research is aimed not only at the promotion of longevity, but also at improving health span through the discovery and development of new therapeutic strategies by investigating molecular and cellular pathways involved in cellular senescence. Understanding the mechanism of action of polyphenolic compounds targeting mTOR (mechanistic target of rapamycin) and related pathways opens up new directions to revolutionize ways to slow down the onset and development of age-dependent degeneration. Herein, we will discuss the mechanisms by which polyphenols can delay the molecular pathogenesis of ageing via manipulation or more specifically inhibition of mTOR-signaling pathways. We will also discuss the implications of polyphenols in targeting mTOR and its related pathways on health life span extension and longevity..
Collapse
|
41
|
Takeshima M, Miyazaki I, Murakami S, Kita T, Asanuma M. l-Theanine protects against excess dopamine-induced neurotoxicity in the presence of astrocytes. J Clin Biochem Nutr 2016. [PMID: 27698535 DOI: 10.3164/jcbn.16.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
l-Theanine (γ-glutamylethylamide), a component of green tea, is considered to have regulatory and neuroprotective roles in the brain. The present study was designed to determine the effect of l-theanine on excess dopamine-induced neurotoxicity in both cell culture and animal experiments. The primary cultured mesencephalic neurons or co-cultures of mesencephalic neurons and striatal astrocytes were pretreated with l-theanine for 72 h, and then treated with excess dopamine for further 24 h. The cell viability of dopamine neurons and levels of glutathione were evaluated. Excess dopamine-induced neurotoxicity was significantly attenuated by 72 h preincubation with l-theanine in neuron-astrocyte co-cultures but not in neuron-rich cultures. Exposure to l-theanine increased the levels of glutathione in both astrocytes and glial conditioned medium. The glial conditioned medium from l-theanine-pretreated striatal astrocytes attenuated dopamine-induced neurotoxicity and quinoprotein formation in mesencephalic neurons. In addition, replacement of l-glutamate with l-theanine in an in vitro cell-free glutathione-synthesis system produced glutathione-like thiol compounds. Furthermore, l-theanine administration (4 mg/kg, p.o.) for 14 days significantly increased glutathione levels in the striatum of mice. The results suggest that l-theanine provides neuroprotection against oxidative stress-induced neuronal damage by humoral molecules released from astrocytes, probably including glutathione.
Collapse
Affiliation(s)
- Mika Takeshima
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Ikuko Miyazaki
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shinki Murakami
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; SAIDO Co., Fukuoka 810-0021, Japan
| | - Taizo Kita
- Laboratory of Pharmacology, Kyushu Nutrition Welfare University School of Health Science, Fukuoka 803-8511, Japan
| | - Masato Asanuma
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
42
|
Takeshima M, Miyazaki I, Murakami S, Kita T, Asanuma M. l-Theanine protects against excess dopamine-induced neurotoxicity in the presence of astrocytes. J Clin Biochem Nutr 2016; 59:93-99. [PMID: 27698535 PMCID: PMC5018574 DOI: 10.3164/jcbn.16-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 04/12/2016] [Indexed: 01/05/2023] Open
Abstract
l-Theanine (γ-glutamylethylamide), a component of green tea, is considered to have regulatory and neuroprotective roles in the brain. The present study was designed to determine the effect of l-theanine on excess dopamine-induced neurotoxicity in both cell culture and animal experiments. The primary cultured mesencephalic neurons or co-cultures of mesencephalic neurons and striatal astrocytes were pretreated with l-theanine for 72 h, and then treated with excess dopamine for further 24 h. The cell viability of dopamine neurons and levels of glutathione were evaluated. Excess dopamine-induced neurotoxicity was significantly attenuated by 72 h preincubation with l-theanine in neuron-astrocyte co-cultures but not in neuron-rich cultures. Exposure to l-theanine increased the levels of glutathione in both astrocytes and glial conditioned medium. The glial conditioned medium from l-theanine-pretreated striatal astrocytes attenuated dopamine-induced neurotoxicity and quinoprotein formation in mesencephalic neurons. In addition, replacement of l-glutamate with l-theanine in an in vitro cell-free glutathione-synthesis system produced glutathione-like thiol compounds. Furthermore, l-theanine administration (4 mg/kg, p.o.) for 14 days significantly increased glutathione levels in the striatum of mice. The results suggest that l-theanine provides neuroprotection against oxidative stress-induced neuronal damage by humoral molecules released from astrocytes, probably including glutathione.
Collapse
Affiliation(s)
- Mika Takeshima
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Ikuko Miyazaki
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shinki Murakami
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; SAIDO Co., Fukuoka 810-0021, Japan
| | - Taizo Kita
- Laboratory of Pharmacology, Kyushu Nutrition Welfare University School of Health Science, Fukuoka 803-8511, Japan
| | - Masato Asanuma
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
43
|
Das J, Singh R, Sharma D. Antiepileptic effect of fisetin in iron-induced experimental model of traumatic epilepsy in rats in the light of electrophysiological, biochemical, and behavioral observations. Nutr Neurosci 2016; 20:255-264. [DOI: 10.1080/1028415x.2016.1183342] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Jharana Das
- Laboratory of Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rameshwar Singh
- Laboratory of Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Deepak Sharma
- Laboratory of Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
44
|
Abstract
Stroke is the second foremost cause of mortality worldwide and a major cause of long-term disability. Due to changes in lifestyle and an aging population, the incidence of stroke continues to increase and stroke mortality predicted to exceed 12 % by the year 2030. However, the development of pharmacological treatments for stroke has failed to progress much in over 20 years since the introduction of the thrombolytic drug, recombinant tissue plasminogen activator. These alarming circumstances caused many research groups to search for alternative treatments in the form of neuroprotectants. Here, we consider the potential use of phytochemicals in the treatment of stroke. Their historical use in traditional medicine and their excellent safety profile make phytochemicals attractive for the development of therapeutics in human diseases. Emerging findings suggest that some phytochemicals have the ability to target multiple pathophysiological processes involved in stroke including oxidative stress, inflammation and apoptotic cell death. Furthermore, epidemiological studies suggest that the consumption of plant sources rich in phytochemicals may reduce stroke risk, and so reinforce the possibility of developing preventative or neuroprotectant therapies for stroke. In this review, we describe results of preclinical studies that demonstrate beneficial effects of phytochemicals in experimental models relevant to stroke pathogenesis, and we consider their possible mechanisms of action.
Collapse
|
45
|
Fisetin and Its Role in Chronic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 928:213-244. [DOI: 10.1007/978-3-319-41334-1_10] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
46
|
Novel water-soluble fisetin/cyclodextrins inclusion complexes: Preparation, characterization, molecular docking and bioavailability. Carbohydr Res 2015; 418:20-28. [PMID: 26531135 DOI: 10.1016/j.carres.2015.09.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/07/2015] [Accepted: 09/25/2015] [Indexed: 12/22/2022]
Abstract
Novel water-soluble inclusion complexes for fisetin (FIT) were developed by introducing β-cyclodextrin (β-CD) and γ-CD. Properties of the obtained complexes, as well as the interactions between each component, were systematically investigated in both solution and solid states by means of ESI-MS, NMR, FT-IR, XRD, DSC, SEM etc. All characterization information demonstrated that FIT/CDs inclusion complexes were formed, and exhibited different spectroscopic features and properties from FIT. A complex with 1:1 stoichiometry of FIT and CDs was confirmed with Job's method. Meanwhile, as supported by molecular modeling calculations, we suggested that phenyl group (C ring) of FIT molecule was included in the CDs cavity from the wide side. Moreover, the water solubility of FIT/CDs was successfully improved from 2.8 mg/mL (in ethanol aqueous solution) to 4.5 mg/mL (FIT/β-CD complex) and 7.8 mg/mL (FIT/γ-CD complex), and higher thermal stability results were shown by thermal analysis for those complexes. Notably, the inclusion complexes displayed almost two times higher cytotoxicity compared to free FIT against Hela and MCF-7 cells. These results suggested that FIT/CDs complexes could be potentially useful in food industry and healthcare area.
Collapse
|
47
|
Zhou CH, Wang CX, Xie GB, Wu LY, Wei YX, Wang Q, Zhang HS, Hang CH, Zhou ML, Shi JX. Fisetin alleviates early brain injury following experimental subarachnoid hemorrhage in rats possibly by suppressing TLR 4/NF-κB signaling pathway. Brain Res 2015; 1629:250-9. [PMID: 26475978 DOI: 10.1016/j.brainres.2015.10.016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 08/13/2015] [Accepted: 10/02/2015] [Indexed: 11/27/2022]
Abstract
Early brain injury (EBI) determines the unfavorable outcomes after subarachnoid hemorrhage (SAH). Fisetin, a natural flavonoid, has anti-inflammatory and neuroprotection properties in several brain injury models, but the role of fisetin on EBI following SAH remains unknown. Our study aimed to explore the effects of fisetin on EBI after SAH in rats. Adult male Sprague-Dawley rats were randomly divided into the sham and SAH groups, fisetin (25mg/kg or 50mg/kg) or equal volume of vehicle was given at 30min after SAH. Neurological scores and brain edema were assayed. The protein expression of toll-like receptor 4 (TLR 4), p65, ZO-1 and bcl-2 was examined by Western blot. TLR 4 and p65 were also assessed by immunohistochemistry (IHC). Enzyme-linked immunosorbent assay (ELISA) was performed to detect the production of pro-inflammatory cytokines. Terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling (TUNEL) was perform to assess neural cell apoptosis. High-dose (50mg/kg) fisetin significantly improved neurological function and reduced brain edema at both 24h and 72h after SAH. Remarkable reductions of TLR 4 expression and nuclear factor κB (NF-κB) translocation to nucleus were detected after fisetin treatment. In addition, fisetin significantly reduced the productions of pro-inflammatory cytokines, decreased neural cell apoptosis and increased the protein expression of ZO-1 and bcl-2. Our data provides the evidence for the first time that fisetin plays a protective role in EBI following SAH possibly by suppressing TLR 4/NF-κB mediated inflammatory pathway.
Collapse
Affiliation(s)
- Chen-hui Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Chun-xi Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Guang-bin Xie
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Ling-yun Wu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Yong-xiang Wei
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Qiang Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Hua-sheng Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Chun-hua Hang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Meng-liang Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Ji-xin Shi
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China.
| |
Collapse
|
48
|
Protective Effect of Edaravone in Primary Cerebellar Granule Neurons against Iodoacetic Acid-Induced Cell Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:606981. [PMID: 26557222 PMCID: PMC4628655 DOI: 10.1155/2015/606981] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/19/2014] [Accepted: 10/22/2014] [Indexed: 01/07/2023]
Abstract
Edaravone (EDA) is clinically used for treatment of acute ischemic stroke in Japan and China due to its potent free radical-scavenging effect. However, it has yet to be determined whether EDA can attenuate iodoacetic acid- (IAA-) induced neuronal death in vitro. In the present study, we investigated the effect of EDA on damage of IAA-induced primary cerebellar granule neurons (CGNs) and its possible underlying mechanisms. We found that EDA attenuated IAA-induced cell injury in CGNs. Moreover, EDA significantly reduced intracellular reactive oxidative stress production, loss of mitochondrial membrane potential, and caspase 3 activity induced by IAA. Taken together, EDA protected CGNs against IAA-induced neuronal damage, which may be attributed to its antiapoptotic and antioxidative activities.
Collapse
|
49
|
Krasieva TB, Ehren J, O'Sullivan T, Tromberg BJ, Maher P. Cell and brain tissue imaging of the flavonoid fisetin using label-free two-photon microscopy. Neurochem Int 2015; 89:243-8. [PMID: 26271433 DOI: 10.1016/j.neuint.2015.08.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 11/18/2022]
Abstract
Over the last few years, we have identified an orally active, novel neuroprotective and cognition-enhancing molecule, the flavonoid fisetin. Fisetin not only has direct antioxidant activity but it can also increase the intracellular levels of glutathione, the major intracellular antioxidant. Fisetin can also activate key neurotrophic factor signaling pathways. In addition, it has anti-inflammatory activity against microglia and astrocytes and inhibits the activity of lipoxygenases, thereby reducing the production of pro-inflammatory eicosanoids and their by-products. However, key questions about its targets and brain penetration remain. In this study, we used label-free two-photon microscopy of intrinsic fisetin fluorescence to examine the localization of fisetin in living nerve cells and the brains of living mice. In cells, fisetin but not structurally related flavonols with different numbers of hydroxyl groups, localized to the nucleoli suggesting that key targets of fisetin may reside in this organelle. In the mouse brain, following intraperitoneal injection and oral administration, fisetin rapidly distributed to the blood vessels of the brain followed by a slower dispersion into the brain parenchyma. Thus, these results provide further support for the effects of fisetin on brain function. In addition, they suggest that label-free two-photon microscopy may prove useful for studying the intracellular and tissue distribution of other intrinsically-fluorescent flavonoids.
Collapse
Affiliation(s)
- Tatiana B Krasieva
- Beckman Laser Institute and Medical Clinic, University of California, 1002 Health Sciences Road, Irvine, CA 92617, USA
| | - Jennifer Ehren
- Cellular Neurobiology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Beckman Laser Institute and Medical Clinic, University of California, 1002 Health Sciences Road, Irvine, CA 92617, USA
| | - Thomas O'Sullivan
- Beckman Laser Institute and Medical Clinic, University of California, 1002 Health Sciences Road, Irvine, CA 92617, USA
| | - Bruce J Tromberg
- Beckman Laser Institute and Medical Clinic, University of California, 1002 Health Sciences Road, Irvine, CA 92617, USA
| | - Pamela Maher
- Cellular Neurobiology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
50
|
Lim JY, Lee JY, Byun BJ, Kim SH. Fisetin targets phosphatidylinositol-3-kinase and induces apoptosis of human B lymphoma Raji cells. Toxicol Rep 2015; 2:984-989. [PMID: 28962438 PMCID: PMC5598213 DOI: 10.1016/j.toxrep.2015.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/29/2015] [Accepted: 07/05/2015] [Indexed: 01/16/2023] Open
Abstract
Fisetin inhibits PI3K activity at the enzymatic and cellular levels. Fisetin induces the apoptosis of Raji cells by downregulating cIAP-2 protein expression. The pro-apoptotic activity of fisetin may be linked to a potential to inhibit mTOR signaling and to induce DNA damage.
Aberrant regulation of phosphatidylinositol-3-kinases (PI3Ks) is known to be involved in the progression of cancers. PI3K-binding flavonoids such as quercetin and myricetin have been shown to inhibit PI3K activity, but the direct targeting of fisetin to PI3K has not been established. Here, we carried out an in silico investigation of fisetin binding to PI3K and determined fisetin’s inhibitory activity in enzymatic and cell-based assays. In addition, fisetin induced apoptosis in human Burkitt’s lymphoma Raji cells by inhibiting both PI3Ks and mammalian target of rapamycin (mTOR). Our results indicate that fisetin may serve as a natural backbone for the development of novel dual inhibitors of PI3Ks and mTOR for the treatment of cancer.
Collapse
Affiliation(s)
- Ji Yeon Lim
- Laboratory of Translational Therapeutics, Korea Research Institute of Chemical Technology, Daejeon 305-600, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Joo Yun Lee
- Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology, Daejeon 305-600, Republic of Korea
| | - Byung Jin Byun
- Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology, Daejeon 305-600, Republic of Korea
- Corresponding author.
| | - Seong Hwan Kim
- Laboratory of Translational Therapeutics, Korea Research Institute of Chemical Technology, Daejeon 305-600, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 305-764, Republic of Korea
- Corresponding author at: Laboratory of Translational Therapeutics, Korea Research Institute of Chemical Technology, Daejeon 305 600, Republic of Korea.
| |
Collapse
|