1
|
Lu YW, Xie LY, Qi MH, Ren S, Wang YQ, Hu JN, Wang Z, Tang S, Zhang JT, Li W. Platycodin D Ameliorates Cognitive Impairment in Type 2 Diabetes Mellitus Mice via Regulating PI3K/Akt/GSK3β Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12516-12528. [PMID: 38491972 DOI: 10.1021/acs.jafc.3c08490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2024]
Abstract
Objectives: The aim of this study was to investigate the ameliorative effect of platycodin D (PD) on cognitive dysfunction in type 2 diabetes mellitus (T2DM) and its potential molecular mechanisms of action in vivo and in vitro. Materials and methods: An animal model of cognitive impairment in T2DM was established using a single intraperitoneal injection of streptozotocin (100 mg/kg) after 8 weeks of feeding a high-fat diet to C57BL/6 mice. In vitro, immunofluorescence staining and Western blot were employed to analyze the effects of PD on glucose-induced neurotoxicity in mouse hippocampal neuronal cells (HT22). Results: PD (2.5 mg/kg) treatment for 4 weeks significantly suppressed the rise in fasting blood glucose in T2DM mice, improved insulin secretion deficiency, and reversed abnormalities in serum triglyceride, cholesterol, low-density lipoprotein, and high-density lipoprotein levels. Meanwhile, PD ameliorated choline dysfunction in T2DM mice and inhibited the production of oxidative stress and apoptosis-related proteins of the caspase family. Notably, PD dose-dependently prevents the loss of mitochondrial membrane potential, promotes phosphorylation of phosphatidylinositol 3 kinase and protein kinase B (Akt) in vitro, activates glycogen synthase kinase 3β (GSK3β) expression at the Ser9 site, and inhibits Tau protein hyperphosphorylation. Conclusions: These findings clearly indicated that PD could alleviate the neurological damage caused by T2DM, and the phosphorylation of Akt at Ser473 may be the key to its effect.
Collapse
Affiliation(s)
- Ya-Wei Lu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Li-Ya Xie
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Meng-Han Qi
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Shen Ren
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Yue-Qi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jun-Nan Hu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Shan Tang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China
- Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Changchun 130118, China
| |
Collapse
|
2
|
Calderaro A, Patanè GT, Tellone E, Barreca D, Ficarra S, Misiti F, Laganà G. The Neuroprotective Potentiality of Flavonoids on Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms232314835. [PMID: 36499159 PMCID: PMC9736131 DOI: 10.3390/ijms232314835] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Alzheimer's disease (AD), due to its spread, has become a global health priority, and is characterized by senile dementia and progressive disability. The main cause of AD and other neurodegenerations (Huntington, Parkinson, Amyotrophic Lateral Sclerosis) are aggregated protein accumulation and oxidative damage. Recent research on secondary metabolites of plants such as polyphenols demonstrated that they may slow the progression of AD. The flavonoids' mechanism of action in AD involved the inhibition of acetylcholinesterase, butyrylcholinesterase, Tau protein aggregation, β-secretase, oxidative stress, inflammation, and apoptosis through modulation of signaling pathways which are implicated in cognitive and neuroprotective functions, such as ERK, PI3-kinase/Akt, NFKB, MAPKs, and endogenous antioxidant enzymatic systems. This review focuses on flavonoids and their role in AD, in terms of therapeutic potentiality for human health, antioxidant potential, and specific AD molecular targets.
Collapse
Affiliation(s)
- Antonella Calderaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Giuseppe Tancredi Patanè
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Ester Tellone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
- Correspondence: (E.T.); (D.B.)
| | - Davide Barreca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
- Correspondence: (E.T.); (D.B.)
| | - Silvana Ficarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Francesco Misiti
- Department of Human Sciences, Society and Health, University of Cassino and Southern Lazio, V. S. Angelo, Loc. Folcara, 3043 Cassino, Italy
| | - Giuseppina Laganà
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
3
|
Xiong Y, Zhou D, Zheng K, Bi W, Dong Y. Extracellular Adenosine Triphosphate Binding to P2Y1 Receptors Prevents Glutamate-Induced Excitotoxicity: Involvement of Erk1/2 Signaling Pathway to Suppress Autophagy. Front Neurosci 2022; 16:901688. [PMID: 35747207 PMCID: PMC9209742 DOI: 10.3389/fnins.2022.901688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/13/2022] [Indexed: 12/20/2022] Open
Abstract
Glutamate-induced neuroexcitotoxicity could be related to the pathophysiology of some neurodegenerative diseases including Parkinson’s disease and Alzheimer’s disease. Extracellular ATP exerts a wide variety of functions, such as attenuating Aβ-mediated toxicity, inhibiting N-Methyl-D-Aspartate (NMDA) receptor subunit combinations, and aggravating ischemic brain injury. However, the effect of extracellular ATP on glutamate-induced neuroexcitotoxicity remains largely unknown. Herein, we showed that extracellular ATP prevented the glutamate-induced excitotoxicity via binding to its P2Y1 receptors. We found that excessive glutamate triggered cellular reactive oxygen species (ROS) overproduction and mitochondrial membrane potential damage, which were significantly attenuated by extracellular ATP. Besides, glutamate activated autophagy, as illustrated by the increased protein level of autophagic marker LC3II and decreased level of p62, and glutamate-induced neuroexcitotoxicity could be completely abolished by autophagy inhibitor chloroquine. In addition, we revealed that extracellular ATP activated Erk1/2 signaling to suppress autophagy and to exert its neuroprotective effects, which was further reduced by autophagy agonist rapamycin and the selective Erk1/2 inhibitor PD0325901. Taken together, our findings suggest that extracellular ATP binding to P2Y1 receptors protected against glutamate-induced excitotoxicity via Erk1/2-mediated autophagy inhibition, implying the potential of ATP for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Yiping Xiong
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Duanyang Zhou
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Kai Zheng
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Wenchuan Bi
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Yun Dong
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
- School of Pharmacy and Food Sciences, Zhuhai College of Science and Technology, Zhuhai, China
- *Correspondence: Yun Dong, ; orcid.org/0000-0002-5658-3896
| |
Collapse
|
4
|
Mak S, Li W, Fu H, Luo J, Cui W, Hu S, Pang Y, Carlier PR, Tsim KW, Pi R, Han Y. Promising tacrine/huperzine A-based dimeric acetylcholinesterase inhibitors for neurodegenerative disorders: From relieving symptoms to modifying diseases through multitarget. J Neurochem 2021; 158:1381-1393. [PMID: 33930191 PMCID: PMC8458250 DOI: 10.1111/jnc.15379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/22/2022]
Abstract
Neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease, are devastating diseases in the elderly world, which are closely associated with progressive neuronal loss induced by a variety of genetic and/or environmental factors. Unfortunately, currently available treatments for neurodegenerative disorders can only relieve the symptoms but not modify the pathological processes. Over the past decades, our group by collaborating with Profs. Yuan-Ping Pang and Paul R. Carlier has developed three series of homo/hetero dimeric acetylcholinesterase inhibitors derived from tacrine and/or huperzine A. The representative dimers bis(3)-Cognitin (B3C), bis(12)-hupyridone, and tacrine(10)-hupyridone might possess disease-modifying effects through the modulation of N-methyl-d-aspartic acid receptors, the activation of myocyte enhancer factor 2D gene transcription, and the promotion of neurotrophic factor secretion. In this review, we summarize that the representative dimers, such as B3C, provide neuroprotection against a variety of neurotoxins via multiple targets, including the inhibitions of N-methyl-d-aspartic acid receptor with pathological-activated potential, neuronal nitric oxide synthase, and β-amyloid cascades synergistically. More importantly, B3C might offer disease-modifying potentials by activating myocyte enhancer factor 2D transcription, inducing neuritogenesis, and promoting the expressions of neurotrophic factors in vitro and in vivo. Taken together, the novel dimers might offer synergistic disease-modifying effects, proving that dimerization might serve as one of the strategies to develop new generation of therapeutics for neurodegenerative disorders.
Collapse
Affiliation(s)
- Shinghung Mak
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China
- Division of Life Science and Center for Chinese Medicine and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Wenming Li
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hongjun Fu
- Department of Neuroscience, Chronic Brain Injury, The Ohio State University, Columbus, OH, USA
| | - Jialie Luo
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Wei Cui
- Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo Key Laboratory of Behavioral Neuroscience, School of Medicine, Ningbo University, Ningbo, China
| | - Shengquan Hu
- Shenzhen Institute of Geriatrics, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yuanping Pang
- Mayo Cancer Center, Department of Pharmacology, Mayo Clinic, Rochester, MN, USA
| | | | - Karl Wahkeung Tsim
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China
- Division of Life Science and Center for Chinese Medicine and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Rongbiao Pi
- Department of Pharmacology, School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology, Institute of Modern Medicine, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
5
|
Wu L, Zhou X, Cao Y, MAK SH, Zha L, Li N, Su Z, Han Y, Wang Y, Man Hoi MP, Sun Y, Zhang G, Zhang Z, Yang X. Therapeutic efficacy of novel memantine nitrate MN-08 in animal models of Alzheimer's disease. Aging Cell 2021; 20:e13371. [PMID: 33955647 PMCID: PMC8208785 DOI: 10.1111/acel.13371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/02/2021] [Accepted: 04/02/2021] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is a leading cause of dementia in elderly individuals and therapeutic options for AD are very limited. Over‐activation of N‐methyl‐D‐aspartate (NMDA) receptors, amyloid β (Aβ) aggregation, a decrease in cerebral blood flow (CBF), and downstream pathological events play important roles in the disease progression of AD. In the present study, MN‐08, a novel memantine nitrate, was found to inhibit Aβ accumulation, prevent neuronal and dendritic spine loss, and consequently attenuate cognitive deficits in 2‐month‐old APP/PS1 transgenic mice (for a 6‐month preventative course) and in the 8‐month‐old triple‐transgenic (3×Tg‐AD) mice (for a 4‐month therapeutic course). In vitro, MN‐08 could bind to and antagonize NMDA receptors, inhibit the calcium influx, and reverse the dysregulations of ERK and PI3K/Akt/GSK3β pathway, subsequently preventing glutamate‐induced neuronal loss. In addition, MN‐08 had favorable pharmacokinetics, blood‐brain barrier penetration, and safety profiles in rats and beagle dogs. These findings suggest that the novel memantine nitrate MN‐08 may be a useful therapeutic agent for AD.
Collapse
Affiliation(s)
- Liangmiao Wu
- Department of Neurology and Stroke Center The First Affiliated Hospital of Jinan University and Institute of New Drug Research International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education Jinan University College of Pharmacy Guangzhou China
| | - Xinhua Zhou
- Department of Neurology and Stroke Center The First Affiliated Hospital of Jinan University and Institute of New Drug Research International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education Jinan University College of Pharmacy Guangzhou China
| | - Yiwan Cao
- Institute of New Drug Research International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education Jinan University College of Pharmacy Guangzhou China
| | - Shing Hung MAK
- Department of Applied Biology and Chemical Technology Institute of Modern Chinese Medicine The Hong Kong Polytechnic University Hong Kong China
| | - Ling Zha
- Institute of New Drug Research International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education Jinan University College of Pharmacy Guangzhou China
| | - Ning Li
- Institute of New Drug Research International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education Jinan University College of Pharmacy Guangzhou China
| | - Zhiyang Su
- Institute of New Drug Research International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education Jinan University College of Pharmacy Guangzhou China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology Institute of Modern Chinese Medicine The Hong Kong Polytechnic University Hong Kong China
| | - Yuqiang Wang
- Institute of New Drug Research International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education Jinan University College of Pharmacy Guangzhou China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences University of Macau Macau China
| | - Yewei Sun
- Institute of New Drug Research International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education Jinan University College of Pharmacy Guangzhou China
| | - Gaoxiao Zhang
- Institute of New Drug Research International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education Jinan University College of Pharmacy Guangzhou China
| | - Zaijun Zhang
- Institute of New Drug Research International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education Jinan University College of Pharmacy Guangzhou China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen Shenzhen Center for Disease Control and Prevention Shenzhen China
| |
Collapse
|
6
|
Wadhwa P, Jain P, Jadhav HR. Glycogen Synthase Kinase 3 (GSK3): Its Role and Inhibitors. Curr Top Med Chem 2021; 20:1522-1534. [PMID: 32416693 DOI: 10.2174/1568026620666200516153136] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 12/23/2022]
Abstract
Glycogen Synthase Kinase 3 (GSK3) is one of the Serine/Threonine protein kinases, which has gained a lot of attention for its role in a variety of pathways. It has two isoforms, GSK3α and GSK3β. However, GSK3β is highly expressed in different areas of the brain and has been implicated in Alzheimer's disease as it is involved in tau phosphorylation. Due to its high specificity concerning substrate recognition, GSK3 has been considered as an important target. In the last decade, several GSK3 inhibitors have been reported and two molecules are in clinical trials. This review collates the information published in the last decade about the role of GSK3 in Alzheimer's disease and progress in the development of its inhibitors. Using this collated information, medicinal chemists can strategize and design novel GSK3 inhibitors that could be useful in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Pankaj Wadhwa
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani- 333031, Rajasthan, India
| | - Priti Jain
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani- 333031, Rajasthan, India
| | - Hemant R Jadhav
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani- 333031, Rajasthan, India
| |
Collapse
|
7
|
Dourado LFN, da Silva FR, Toledo CR, da Silva CN, Santana CP, da Costa BL, de Lima ME, Cunha ADS. Intravitreal injection of peptides PnPa11 and PnPa13, derivatives of Phoneutria nigriventer spider venom, prevents retinal damage. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20200031. [PMID: 33014024 PMCID: PMC7518191 DOI: 10.1590/1678-9199-jvatitd-2020-0031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 08/18/2020] [Indexed: 12/18/2022] Open
Abstract
Background: PnPa11 and PnPa13 are synthetic peptides derived from Phoneutria
nigriventer spider venom, which display antinociceptive and
neuroprotective properties. In this work, we evaluated the safety of
intravitreal use and the neuroprotective effect of these peptides. Methods: The cytotoxicity and the antiangiogenic activity of these peptides were
evaluated by the sulforhodamine-B method and chicken chorioallantoic
membrane (CAM) assay, respectively. The in vivo safety was
analyzed in Wistar rats that were intravitreally injected with different
doses (0.50; 1.25; 2.50; 3.75 and 5.00 µg/mL) of these peptides (right eye,
n = 6). The retinal function was assessed by electroretinography exams
(ERG), intraocular pressure (IOP), and histological analyzes. In order to
investigate the neuroprotective effect, Wistar rats received intravitreal
injections (right eye, n = 6) of peptides at 1.25 µg/mL and then were
exposed to blue LED light. In addition, the visual function and the retinal
microstructure were verified. Results: Cytotoxicity analyses demonstrated that the peptides did not present any
toxicity over ARPE-19 (adult retinal pigmented epithelial) cell line and the
antiangiogenic study highlighted that the peptides promoted the reduction of
blood vessels. The intravitreal injection did not cause major changes,
neither induced any irreversible damage. In the retinal degeneration assay,
the ERG records demonstrated that the prior treatment with PnPa11 and PnPa13
protected the retina from damage. Morphological analyses confirmed the ERG
findings. Immunoblotting analyses revealed that PnPa11 increased Erk1/2,
NR2A, and NR2B retinal expression after the light stress model, but did not
cause Akt1 activation, while PnPa13 prevented Erk1/2 and Akt1
dephosphorylation. Conclusions: The intraocular administration of these peptides was well tolerated and
presented protective activity against retinal degeneration, suggesting the
potential use of these peptides as neuroprotectors in the ophthalmological
field.
Collapse
Affiliation(s)
| | - Flavia Rodrigues da Silva
- School of Pharmacy, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.,National Institute of Science and Technology in Pharmaceutical Nanotechnology, São Paulo, SP, Brazil
| | | | | | | | - Bruna Lopes da Costa
- School of Pharmacy, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Maria Elena de Lima
- Graduate Program in Health Sciences: Medicine and Biomedicine, Institute of Education and Research, Santa Casa de Belo Horizonte, Belo Horizonte, MG, Brazil
| | - Armando da Silva Cunha
- School of Pharmacy, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
8
|
Gerace E, Scartabelli T, Pellegrini-Giampietro DE, Landucci E. Tolerance Induced by (S)-3,5-Dihydroxyphenylglycine Postconditioning is Mediated by the PI3K/Akt/GSK3β Signalling Pathway in an In Vitro Model of Cerebral Ischemia. Neuroscience 2020; 433:221-229. [DOI: 10.1016/j.neuroscience.2019.12.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/13/2019] [Accepted: 12/30/2019] [Indexed: 12/16/2022]
|
9
|
Liu Z, Qiu X, Mak S, Guo B, Hu S, Wang J, Luo F, Xu D, Sun Y, Zhang G, Cui G, Wang Y, Zhang Z, Han Y. Multifunctional memantine nitrate significantly protects against glutamate-induced excitotoxicity via inhibiting calcium influx and attenuating PI3K/Akt/GSK3beta pathway. Chem Biol Interact 2020; 325:109020. [PMID: 32092300 DOI: 10.1016/j.cbi.2020.109020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 10/09/2019] [Accepted: 02/18/2020] [Indexed: 01/30/2023]
Abstract
Overactivation of N-methyl-D-aspartate (NMDA) receptors has been associated with neurodegenerative disorders such as Alzheimer's disease (AD), cerebral vascular disorders and amyotrophic lateral sclerosis (ALS). We have previously designed and synthesized a series of memantine nitrate and some of them have shown vessel dilatory effects and neuroprotective effects; however, the detailed mechanisms have not been elucidated. In this study, we further demonstrated that memantine nitrate-06 (MN-06), one of the novel compounds derived from memantine, possessed significant neuroprotective effects against glutamate-induced excitotoxicity in rat primary cerebellar granule neurons (CGNs). Pretreatment of MN-06 reversed the activation of GSK3b and the suppression of phosphorylated Akt induced by glutamate. In addition, the neuroprotective effects of MN-06 could be abolished by LY294002, the specific phosphatidylinositol 3-kinase (PI3-K) inhibitor. Ca2+ imaging shown that pretreatment of MN-06 prevented Ca2+ influx induced by glutamate. Moreover, MN-06 might inhibit the NMDA-mediated current by antagonizing NDMA receptors, which was further confirmed by molecular docking simulation. Taken together, MN-06 protected against glutamate-induced excitotoxicity by blocking calcium influx and attenuating PI3-K/Akt/GSK-3b pathway, indicating that MN-06 might be a potential drug for treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Zheng Liu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Xiaoling Qiu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Shinghung Mak
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China; Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Baojian Guo
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Shengquan Hu
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China; Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Jiajun Wang
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China; Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Fangcheng Luo
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Daping Xu
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China; Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Yewei Sun
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Gaoxiao Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Guozhen Cui
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, China
| | - Yuqiang Wang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China.
| | - Yifan Han
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China; Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China.
| |
Collapse
|
10
|
Mak S, Liu Z, Wu L, Guo B, Luo F, Liu Z, Hu S, Wang J, Cui G, Sun Y, Wang Y, Zhang G, Han Y, Zhang Z. Pharmacological Characterizations of anti-Dementia Memantine Nitrate via Neuroprotection and Vasodilation in Vitro and in Vivo. ACS Chem Neurosci 2020; 11:314-327. [PMID: 31922720 DOI: 10.1021/acschemneuro.9b00242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We have previously designed and synthesized a series of novel memantine nitrates, and some of them have shown neuroprotective effects; however, the detailed mechanisms remain unknown. In this study, we demonstrated that MN-12, one of the memantine nitrates, concentration-dependently protected against glutamate-induced neurotoxicity in rat primary cultured cerebellar granule neurons (CGNs). Western blotting assays revealed that MN-12 might possess neuroprotective effects through the inhibition of ERK pathway and activation of PI3K/Akt pathway concurrently. Moreover, MN-12 concentration-dependently dilated precontracted rat middle cerebral artery through activation of NO-cGMP pathway ex vivo. In the 2-vessel occlusion (2VO) rat model, MN-12 alleviated the impairments of spatial memory and motor dysfunction possibly via neuroprotection and improvement of the cerebral blood flow. Furthermore, the results of preliminary pharmacokinetic studies showed that MN-12 might quickly distribute to the major organs including the brain, indicating that MN-12 could penetrate the blood-brain barrier. Taken together, MN-12 might provide multifunctional therapeutic benefits for dementia associated with Alzheimer's disease, vascular dementia, and ischemic stroke, via neuroprotection and vessel dilation to improve the cerebral blood flow.
Collapse
Affiliation(s)
- Shinghung Mak
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation) , The Hong Kong Polytechnic University Shenzhen Research Institute , Shenzhen 518057 , China
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine , The Hong Kong Polytechnic University , Hung Hom, Hong Kong , China
| | - Zheng Liu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases , Jinan University College of Pharmacy , Guangzhou 510632 , China
- Foshan Stomatology Hospital, School of Stomatology and Medicine , Foshan University , Foshan 528000 , China
- Foshan Magpie Pharmaceuticals Co., Ltd. , Foshan , 528000 Guangdong , China
| | - Liangmiao Wu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases , Jinan University College of Pharmacy , Guangzhou 510632 , China
| | - Baojian Guo
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation) , The Hong Kong Polytechnic University Shenzhen Research Institute , Shenzhen 518057 , China
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases , Jinan University College of Pharmacy , Guangzhou 510632 , China
| | - Fangcheng Luo
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases , Jinan University College of Pharmacy , Guangzhou 510632 , China
- Foshan Magpie Pharmaceuticals Co., Ltd. , Foshan , 528000 Guangdong , China
| | - Ziyan Liu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases , Jinan University College of Pharmacy , Guangzhou 510632 , China
| | - Shengquan Hu
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation) , The Hong Kong Polytechnic University Shenzhen Research Institute , Shenzhen 518057 , China
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine , The Hong Kong Polytechnic University , Hung Hom, Hong Kong , China
| | - Jiajun Wang
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine , The Hong Kong Polytechnic University , Hung Hom, Hong Kong , China
| | - Guozhen Cui
- Department of Bioengineering , Zunyi Medical University Zhuhai Campus , Zhuhai 519041 , China
| | - Yewei Sun
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases , Jinan University College of Pharmacy , Guangzhou 510632 , China
| | - Yuqiang Wang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases , Jinan University College of Pharmacy , Guangzhou 510632 , China
| | - Gaoxiao Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases , Jinan University College of Pharmacy , Guangzhou 510632 , China
| | - Yifan Han
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation) , The Hong Kong Polytechnic University Shenzhen Research Institute , Shenzhen 518057 , China
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine , The Hong Kong Polytechnic University , Hung Hom, Hong Kong , China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases , Jinan University College of Pharmacy , Guangzhou 510632 , China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE) , Jinan University College of Pharmacy , 601 Huangpu Avenue West , Guangzhou 510632 , China
| |
Collapse
|
11
|
Zuo X, Hu S, Tang Y, Zhan L, Sun W, Zheng J, Han Y, Xu E. Attenuation of secondary damage and Aβ deposits in the ipsilateral thalamus of dMCAO rats through reduction of cathepsin B by bis(propyl)-cognitin, a multifunctional dimer. Neuropharmacology 2020; 162:107786. [DOI: 10.1016/j.neuropharm.2019.107786] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 09/01/2019] [Accepted: 09/19/2019] [Indexed: 10/25/2022]
|
12
|
Jia L, Wang W, Sang J, Wei W, Zhao W, Lu F, Liu F. Amyloidogenicity and Cytotoxicity of a Recombinant C-Terminal His 6-Tagged Aβ 1-42. ACS Chem Neurosci 2019; 10:1251-1262. [PMID: 30537813 DOI: 10.1021/acschemneuro.8b00333] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aggregation of amyloid β peptide (Aβ) is closely associated with the occurrence and development of Alzheimer's disease (AD). Reproducible and detailed studies on the aggregation kinetics and structure of various aggregates have been conducted using recombinant Aβ peptides. While the His6-tag is commonly used in the purification of recombinant proteins due to its great simplicity and affinity, there is little information on the aggregation of His6-tagged Aβ and its corresponding cytotoxicity. Moreover, it is also unclear whether there is an effect of the His6-tag on the amyloidogenicity and cytotoxicity of recombinant Aβ1-42. Herein, a method to express and purify a mutant C-terminally His6-tagged Aβ1-42 (named as Aβ1-42-His6) from Escherichia coli was described. Aβ1-42-His6 aggregated into β-sheet-rich fibrils as shown by thioflavin T fluorescence, atomic force microscopy and circular dichroism spectroscopy. Moreover, the fibrillar recombinant Aβ1-42-His6 showed strong toxicity toward PC12 cells in vitro. Molecular dynamics simulations revealed that the His6-tag contributed little to the secondary structure and intermolecular interactions, including hydrophobic interactions, salt bridges, and hydrogen bonding of the fibrillar pentamer of Aβ1-42. This highlights the biological importance of modification on the molecular structure of Aβ. Thus, the easily purified high-quality Aβ1-42-His6 offers great advantages for screening aggregation inhibitors or in vitro confirmation of rationally designed drugs for the treatment of AD.
Collapse
Affiliation(s)
- Longgang Jia
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Wenjuan Wang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Jingcheng Sang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Wei Wei
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Wenping Zhao
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Fuping Lu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Fufeng Liu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| |
Collapse
|
13
|
Neuroprotective effects of cordycepin inhibit Aβ-induced apoptosis in hippocampal neurons. Neurotoxicology 2018; 68:73-80. [DOI: 10.1016/j.neuro.2018.07.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/26/2018] [Accepted: 07/15/2018] [Indexed: 12/12/2022]
|
14
|
Chen H, Xiang S, Huang L, Lin J, Hu S, Mak SH, Wang C, Wang Q, Cui W, Han Y. Tacrine(10)-hupyridone, a dual-binding acetylcholinesterase inhibitor, potently attenuates scopolamine-induced impairments of cognition in mice. Metab Brain Dis 2018; 33:1131-1139. [PMID: 29564727 DOI: 10.1007/s11011-018-0221-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 03/15/2018] [Indexed: 01/08/2023]
Abstract
Tacrine(10)-hupyridone (A10E) was designed as a dual-binding acetylcholinesterase (AChE) inhibitor from the modification of tacrine and a fragment of huperzine A. We have found that A10E effectively inhibited AChE in a mixed competitive manner, with an IC50 of 26.4 nM, which is more potent than those of tacrine and huperzine A. Most importantly, we have shown, for the first time that A10E attenuated scopolamine-induced cognitive impairments without affecting motor function in mice. A10E effectively attenuated impairments of learning and memory to a similar extent as donepezil, an inhibitor of AChE used for treating Alzheimer's disease (AD). In addition, A10E significantly decreased AChE activity in the brain of mice, suggesting that A10E might cross the brain blood-barrier. Taken together, our results demonstrated that A10E, a designed dual-binding AChE inhibitor, could effectively reverse cognitive impairments, indicating that A10E might provide therapeutic efficacy for AD treatment.
Collapse
Affiliation(s)
- Huixin Chen
- Research Center of Behavioural Science, Department of Physiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Siying Xiang
- Research Center of Behavioural Science, Department of Physiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Ling Huang
- Research Center of Behavioural Science, Department of Physiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Jiajia Lin
- Research Center of Behavioural Science, Department of Physiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Shengquan Hu
- Department of Applied Biology and Chemistry Technology, Institute of Modern Chinese Medicine, the Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Shing-Hung Mak
- Department of Applied Biology and Chemistry Technology, Institute of Modern Chinese Medicine, the Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Chuang Wang
- Research Center of Behavioural Science, Department of Physiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Qinwen Wang
- Research Center of Behavioural Science, Department of Physiology, School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Wei Cui
- Research Center of Behavioural Science, Department of Physiology, School of Medicine, Ningbo University, Ningbo, 315211, China.
| | - Yifan Han
- Department of Applied Biology and Chemistry Technology, Institute of Modern Chinese Medicine, the Hong Kong Polytechnic University, Hung Hom, Hong Kong, China.
| |
Collapse
|
15
|
Hu S, Mak S, Zuo X, Li H, Wang Y, Han Y. Neuroprotection Against MPP +-Induced Cytotoxicity Through the Activation of PI3-K/Akt/GSK3β/MEF2D Signaling Pathway by Rhynchophylline, the Major Tetracyclic Oxindole Alkaloid Isolated From Uncaria rhynchophylla. Front Pharmacol 2018; 9:768. [PMID: 30072894 PMCID: PMC6060423 DOI: 10.3389/fphar.2018.00768] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/25/2018] [Indexed: 11/13/2022] Open
Abstract
Rhynchophylline is a major tetracyclic oxindole alkaloid in Uncaria rhynchophylla, which has been extensively used as traditional herb medicine for the prevention of convulsions and hypertension. However, there is still little evidence about the neuroprotective effects of rhynchophylline for Parkinson’s disease (PD), a neurodegenerative condition currently without any effective cure. In this present study, the neuroprotective molecular mechanisms of rhynchophylline were investigated in a cellular model associated with PD. It is shown that rhynchophylline (10–50 μM) greatly prevented neurotoxicity caused by 1-methyl-4-phenylpyridinium ion (MPP+) in primary cerebellar granule neurons, as evidenced by the promotion of cell viability as well as the reversal of dysregulated protein expression of Bax/Bcl-2 ratio. Very encouragingly, we found that rhynchophylline markedly enhanced the activity of the transcription factor myocyte enhancer factor 2D (MEF2D) at both basal and pathological conditions using luciferase reporter gene assay, and reversed the inhibition of MEF2D caused by MPP+. Additionally, pharmacological inhibition of PI3-Kinase or short hairpin RNA-mediated gene down-regulation of MEF2D abrogated the protection provided by rhynchophylline. Furthermore, Western blot analysis revealed that rhynchophylline could potentiate PI3-K/Akt to attenuate GSK3β (the MEF2D inhibitor) in response to MPP+ insult. In conclusion, rhynchophylline inhibits MPP+-triggered neurotoxicity by stimulating MEF2D via activating PI3-K/Akt/GSK3β cascade. Rhynchophylline is served as a novel MEF2D enhancer and might be a potential candidate for further preclinical study in the prevention of PD.
Collapse
Affiliation(s)
- Shengquan Hu
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.,Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Shinghung Mak
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.,Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Xialin Zuo
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Haitao Li
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Yuqiang Wang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Yifan Han
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.,Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| |
Collapse
|
16
|
Hu S, Hu H, Mak S, Cui G, Lee M, Shan L, Wang Y, Lin H, Zhang Z, Han Y. A Novel Tetramethylpyrazine Derivative Prophylactically Protects against Glutamate-Induced Excitotoxicity in Primary Neurons through the Blockage of N-Methyl-D-aspartate Receptor. Front Pharmacol 2018; 9:73. [PMID: 29483871 PMCID: PMC5816056 DOI: 10.3389/fphar.2018.00073] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/22/2018] [Indexed: 12/18/2022] Open
Abstract
The over-activation of NMDA receptor via the excessive glutamate is believed to one of the most causal factors associated with Alzheimer’s disease (AD), a progressive neurodegenerative brain disorder. Molecules that could protect against glutamate-induced neurotoxicity may hold therapeutic values for treating AD. Herein, the neuroprotective mechanisms of dimeric DT-010, a novel derivative of naturally occurring danshensu and tetramethylpyrazine, were investigated using primary rat cerebellar granule neurons (CGNs) and hippocampal neurons. It was found that DT-010 (3–30 μM) markedly prevented excitotoxicity of CGNs caused by glutamate, as evidenced by the promotion of neuronal viability as well as the reversal of abnormal morphological changes. While its parent molecules did not show any protective effects even when their concentration reached 50 μM. Additionally, DT-010 almost fully blocked intracellular accumulation of reactive oxygen species caused by glutamate and exogenous oxidative stimulus. Moreover, Western blot results demonstrated that DT-010 remarkably attenuated the inhibition of pro-survival PI3K/Akt/GSK3β pathway caused by glutamate. Ca2+ imaging with Fluo-4 fluorescence analysis further revealed that DT-010 greatly declined glutamate-induced increase in intracellular Ca2+. Most importantly, with the use of whole-cell patch clamp electrophysiology, DT-010 directly inhibited NMDA-activated whole-cell currents in primary hippocampal neurons. Molecular docking simulation analysis further revealed a possible binding mode that inhibited NMDA receptor at the ion channel, showing that DT-010 favorably binds to Asn602 of NMDA receptor via arene hydrogen bond. These results suggest that DT-010 could be served as a novel NMDA receptor antagonist and protect against glutamate-induced excitotoxicity from blocking the upstream NMDA receptors to the subsequent Ca2+ influx and to the downstream GSK3β cascade.
Collapse
Affiliation(s)
- Shengquan Hu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China.,Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hong Kong, China
| | - Huihui Hu
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Shinghung Mak
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hong Kong, China.,Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen, China
| | - Guozhen Cui
- Department of Bioengineering, Zunyi Medical University, Zhuhai, China
| | - Mingyuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| | - Luchen Shan
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Yuqiang Wang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Huangquan Lin
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hong Kong, China.,Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen, China
| |
Collapse
|
17
|
Jang S, Park SH. Antidiabetic Drug Metformin Protects Neuronal Cells against Quinolinic Acid-Induced Excitotoxicity by Decreasing Intracellular Calcium. Chonnam Med J 2018; 54:24-30. [PMID: 29399562 PMCID: PMC5794475 DOI: 10.4068/cmj.2018.54.1.24] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/19/2018] [Accepted: 01/20/2018] [Indexed: 12/19/2022] Open
Abstract
The antidiabetic drug metformin has been found to have beneficial effects in various neurological disorders; however, the molecular mechanisms underlying these effects remain unclear. Here we report that metformin protects neuronal cells from quinolinic acid (QUIN)-induced excitotoxicity. For this, we pretreated N18D3 neuronal cells with metformin prior to QUIN for 24 h. We found that pretreating the cells with metformin significantly improved cell survival rate in a concentration-dependent manner and reduced apoptotic cell death, as revealed by a MTT assay and DAPI staining, respectively. Calcium imaging using fluo-4 showed that metformin (100 µM) inhibited the intracellular calcium increase that was induced by QUIN. In addition, mRNA expression of pro-apoptotic genes, p21 and Bax, was decreased and of anti-apoptotic genes, Bcl-2 and Bcl-xl, was increased with metformin treatment compared to QUIN-induced cells. The immunoreactivity of phosphorylated ERK1/2 was elevated in cells treated with metformin, indicating the ERK1/2 signaling pathway in the neuroprotective effects of metformin in QUIN-induced cell death. Collectively, our data demonstrates that metformin exerts its neuroprotective effects by inhibiting intracellular calcium increases, allowing it to regulate ERK1/2 signaling and modulate cell survival and death genes.
Collapse
Affiliation(s)
- Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Gwangju, Korea.,Research Institute of Medical Sciences, Chonnam National University, Gwangju, Korea
| | - Sah-Hoon Park
- Department of Physiology, Chonnam National University Medical School, Gwangju, Korea.,Research Institute of Medical Sciences, Chonnam National University, Gwangju, Korea
| |
Collapse
|
18
|
Bali A, Jaggi AS. Angiotensin II-triggered kinase signaling cascade in the central nervous system. Rev Neurosci 2018; 27:301-15. [PMID: 26574890 DOI: 10.1515/revneuro-2015-0041] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 09/26/2015] [Indexed: 12/26/2022]
Abstract
Recent studies have projected the renin-angiotensin system as a central component of the physiological and pathological processes of assorted neurological disorders. Its primary effector hormone, angiotensin II (Ang II), not only mediates the physiological effects of vasoconstriction and blood pressure regulation in cardiovascular disease but is also implicated in a much wider range of neuronal activities and diseases, including Alzheimer's disease, neuronal injury, and cognitive disorders. Ang II produces different actions by acting on its two subtypes of receptors (AT1 and AT2); however, the well-known physiological actions of Ang II are mainly mediated through AT1 receptors. Moreover, recent studies also suggest the important functional role of AT2 receptor in the brain. Ang II acts on AT1 receptors and conducts its functions via MAP kinases (ERK1/2, JNK, and p38MAPK), glycogen synthase kinase, Rho/ROCK kinase, receptor tyrosine kinases (PDGF and EGFR), and nonreceptor tyrosine kinases (Src, Pyk2, and JAK/STAT). AT1R-mediated NADPH oxidase activation also leads to the generation of reactive oxygen species, widely implicated in neuroinflammation. These signaling cascades lead to glutamate excitotoxicity, apoptosis, cerebral infarction, astrocyte proliferation, nociception, neuroinflammation, and progression of other neurological disorders. The present review focuses on the Ang II-triggered signal transduction pathways in central nervous system.
Collapse
|
19
|
Deng LJ, Cheng C, Wu J, Wang CH, Zhou HB, Huang J. Oxabicycloheptene Sulfonate Protects Against β-Amyloid-induced Toxicity by Activation of PI3K/Akt and ERK Signaling Pathways Via GPER1 in C6 Cells. Neurochem Res 2017; 42:2246-2256. [DOI: 10.1007/s11064-017-2237-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/20/2017] [Accepted: 03/14/2017] [Indexed: 10/19/2022]
|
20
|
|
21
|
Xu W, Yang L, Li J. Protection against β-amyloid-induced neurotoxicity by naturally occurring Z-ligustilide through the concurrent regulation of p38 and PI3-K/Akt pathways. Neurochem Int 2016; 100:44-51. [PMID: 27580711 DOI: 10.1016/j.neuint.2016.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/12/2016] [Accepted: 08/26/2016] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease (AD) is primarily characterized by the progressive loss of functional neurons in the brain. Therefore, compounds with neuroprotective property may have therapeutic value in treating AD. Z-ligustilide (Z-LIG) is an essential oil originally isolated from umbelliferous plants. In the current study, the neuroprotective effects and underlying mechanisms of Z-LIG against fibrillar aggregates of Aβ25-35 and Aβ1-42-induced neurotoxicity were investigated in both SH-SY5Y cells and differentiated PC12 cells. Z-LIG at 1-30 μM provided an effective neuroprotection, as evidenced by the increase in cell viability, as well as the decrease in LDH release and intracellular accumulation of reactive oxygen species. Additionally, Z-LIG markedly blocked Aβ fibrils-induced condensed nuclei and sub-G1 accumulation suggestive of apoptosis. Furthermore, Z-LIG substantially reversed the activation of phosphorylated p38 and the inhibition of phosphorylated Akt caused by Aβ25-35. LY294002, the specific inhibitor of PI3-K, significantly abrogated the protein expression of up-regulated phosphorylated Akt offered by Z-LIG. Most importantly, siRNA-mediated knockdown of PI3-K and p38 significantly abolished the neuroprotective effects of Z-LIG. The results taken together indicate that Z-LIG protects against Aβ fibrils-induced neurotoxicity possibly through the inhibition of p38 and activation of PI3-K/Akt signaling pathways concurrently. Z-LIG might be a potential candidate for further preclinical study aimed at the prevention and treatment of AD.
Collapse
Affiliation(s)
- Wei Xu
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.
| | - Li Yang
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Ji Li
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
22
|
Neuroprotection against glutamate-induced excitotoxicity and induction of neurite outgrowth by T-006, a novel multifunctional derivative of tetramethylpyrazine in neuronal cell models. Neurochem Int 2016; 99:194-205. [PMID: 27445088 DOI: 10.1016/j.neuint.2016.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 07/05/2016] [Accepted: 07/13/2016] [Indexed: 01/04/2023]
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder, characterized by irreversible impairment of memory and cognitive function. The exact causes of Alzheimer's disease still remain unclear and current single target drugs could only offer limited therapeutic effect to the patients. We have previously reported that T-006, a promising anti-Alzheimer's compound derived from Chinese medicinal component tetramethylpyrazine, might protect neurons through inhibiting the overproduction of intracellular reactive oxygen species (ROS) and reactive nitrogen species (RNS). In this study, we further investigated the neuroprotective effects, as well as the molecular pathways involved, of T-006 against glutamate-induced excitotoxicity in rat cerebellar granule neurons (CGNs). T-006 was also found to promote neuronal differentiation in both PC12 cells and primary cultured rat cortical neurons. The results showed that the pretreatment of T-006 (0.01-1 μM) might prevent glutamate-induced neuronal loss in a concentration-dependent manner. T-006 is found to inhibit the over-activation of NMDAR and ensued calcium overload caused by glutamate. The following activation of phosphorylated extracellular signal-regulated kinase (ERK) were also abolished. Moreover, T-006 concurrently prevented the suppression of phosphorylated protein kinase B (Akt) and glycogen synthase kinase 3β (GSK3β). T-006 was also found to promote neurite outgrowth in PC12 cells and primary cortical neurons. In our study, T-006 (0.1-3 μM) dose-dependently stimulated neurite outgrowth in PC12 cells and the efficacy was comparable to nerve growth factor (NGF). Moreover, co-treatment of T-006 and NGF revealed that T-006 could robustly potentiate the NGF-induced neuritogenesis. Further signal transduction studies indicated that T-006 rapidly up-regulated phosphorylation of ERK but did not activate tyrosine kinase receptor A (Trk A). These findings offer deeper understanding of the anti-neurodegenerative activity of T-006 and provide insight into its possible therapeutic potential for AD treatment in light of the multipotent nature of T-006.
Collapse
|
23
|
Potent Protection Against MPP +-Induced Neurotoxicity via Activating Transcription Factor MEF2D by a Novel Derivative of Naturally Occurring Danshensu/Tetramethylpyrazine. Neuromolecular Med 2016; 18:561-572. [PMID: 27277280 DOI: 10.1007/s12017-016-8399-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/21/2016] [Indexed: 01/05/2023]
Abstract
Danshensu (DSS) and tetramethylpyrazine (TMP) are active ingredients of Salvia miltiorrhiza Bge. and Ligusticum chuanxiong Hort that are widely used in oriental medicine. Structural combination of compounds with known biological activity may lead to the formation of a molecule with multiple properties or new function profile. In the current study, the neuroprotective effects of DT-010, a novel analogue in which TMP was coupled to DSS through an ester bond and two allyl groups at the carboxyl group, were evaluated in a cellular model of Parkinson's disease (PD). As evidenced by the increase in cell survival, as well as the decrease in the number of Hoechst-stained apoptotic nuclei and the level of intracellular accumulation of reactive oxygen species, DT-010 at 3-30 µM substantially protected against MPP+-induced neurotoxicity in both PC12 cells and primary cerebellar granule neurons, a protection that was more potent and efficacious than its parent molecules DSS and TMP. Very encouragingly, we found that DT-010, but not DSS or TMP, could enhance myocyte enhancer factor 2D (MEF2D) transcriptional activity using luciferase reporter gene assay. The neuroprotective effects of DT-010 could be blocked by pharmacologic inhibition of PI3K pathways with LY294002, or MEF2D pathway with short hairpin RNA-mediated knockdown of MEF2D. Furthermore, western blot analysis revealed that DT-010 potentiates Akt protein expression against MPP+ to down-regulate MEF2D inhibitor GSK3β. Taken together, the results suggest that DT-010 prevents MPP+-induced neurotoxicity via enhancing MEF2D through the activation of PI3K/Akt/GSK3β pathway. DT-010 may be a potential candidate for further preclinical study for preventing and treating PD.
Collapse
|
24
|
Jiang W, Luo T, Li S, Zhou Y, Shen XY, He F, Xu J, Wang HQ. Quercetin Protects against Okadaic Acid-Induced Injury via MAPK and PI3K/Akt/GSK3β Signaling Pathways in HT22 Hippocampal Neurons. PLoS One 2016; 11:e0152371. [PMID: 27050422 PMCID: PMC4822954 DOI: 10.1371/journal.pone.0152371] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 03/14/2016] [Indexed: 11/28/2022] Open
Abstract
Increasing evidence shows that oxidative stress and the hyperphosphorylation of tau protein play essential roles in the progression of Alzheimer’s disease (AD). Quercetin is a major flavonoid that has anti-oxidant, anti-cancer and anti-inflammatory properties. We investigated the neuroprotective effects of quercetin to HT22 cells (a cell line from mouse hippocampal neurons). We found that Okadaic acid (OA) induced the hyperphosphorylation of tau protein at Ser199, Ser396, Thr205, and Thr231 and produced oxidative stress to the HT22 cells. The oxidative stress suppressed the cell viability and decreased the levels of lactate dehydrogenase (LDH), superoxide dismutase (SOD), mitochondria membrane potential (MMP) and Glutathione peroxidase (GSH-Px). It up-regulated malondialdehyde (MDA) production and intracellular reactive oxygen species (ROS). In addition, phosphoinositide 3 kinase/protein kinase B/Glycogen synthase kinase3β (PI3K/Akt/GSK3β) and mitogen activated protein kinase (MAPK) were also involved in this process. We found that pre-treatment with quercetin can inhibited OA-induced the hyperphosphorylation of tau protein and oxidative stress. Moreover, pre-treatment with quercetin not only inhibited OA-induced apoptosis via the reduction of Bax, and up-regulation of cleaved caspase 3, but also via the inhibition of PI3K/Akt/GSK3β, MAPKs and activation of NF-κB p65. Our findings suggest the therapeutic potential of quercetin to treat AD.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Department of Anatomy and Histoembryology, Zhaoqing Medical College, Zhaoqing, Guangdong, 526020, China
| | - Tao Luo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Sheng Li
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yue Zhou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiu-Yin Shen
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Feng He
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
- * E-mail: (FH); (HQW)
| | - Jie Xu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Hua-Qiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- * E-mail: (FH); (HQW)
| |
Collapse
|
25
|
Zhao Q, Ye J, Wei N, Fong C, Dong X. Protection against MPP(+)-induced neurotoxicity in SH-SY5Y cells by tormentic acid via the activation of PI3-K/Akt/GSK3β pathway. Neurochem Int 2016; 97:117-23. [PMID: 26994872 DOI: 10.1016/j.neuint.2016.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 03/08/2016] [Accepted: 03/14/2016] [Indexed: 01/08/2023]
Abstract
The cause of Parkinson's disease (PD) could be ascribed to the progressive and selective loss of dopaminergic neurons in the substantia nigra pars compacta, and thus molecules with neuroprotective ability may have therapeutic value against PD. In the current study, the neuroprotective effects and underlying mechanisms of tormentic acid (TA), a naturally occurring triterpene extracted from medicinal plants such as Rosa rugosa and Potentilla chinensis, were evaluated in a widely used cellular PD model in which neurotoxicity was induced by MPP(+) in cultured SH-SY5Y cells. We found that TA at 1-30 μM substantially protected against MPP(+)-induced neurotoxicity, as evidenced by the increase in cell viability, decrease in lactate dehydrogenase release and the reduction in apoptotic nuclei. Moreover, TA effectively inhibited the elevated intracellular accumulation of reactive oxygen species as well as Bax/Bcl-2 ratio caused by MPP(+). Most importantly, TA markedly reversed the inhibition of protein expression of phosphorylated Akt (Ser 473) and phosphorylated GSK3β (Ser 9) caused by MPP(+). LY294002, the specific inhibitor of PI3-K, significantly abrogated the up-regulated phosphorylated Akt and phosphorylated GSK3β offered by TA, suggesting that the neuroprotection of TA was mainly dependent on the activation of PI3-K/Akt/GSK3β signaling pathway. The results taken together indicate that TA may be a potential candidate for further preclinical study aimed at the prevention and treatment of PD.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Neurology, Linzi Maternal & Child Health Hospital of Zibo, Zibo, Shandong, China
| | - Junli Ye
- Department of Pathophysiology, Medical College, Qingdao University, Qingdao, Shandong, China
| | - Na Wei
- Department of Neurology, Linzi Maternal & Child Health Hospital of Zibo, Zibo, Shandong, China
| | - Chichun Fong
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Xiaoli Dong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China; Shenzhen Research Institute of the Hong Kong Polytechnic University, State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen, Guangdong, China.
| |
Collapse
|
26
|
Gao Y, Xu X, Chang S, Wang Y, Xu Y, Ran S, Huang Z, Li P, Li J, Zhang L, Saavedra JM, Liao H, Pang T. Totarol prevents neuronal injury in vitro and ameliorates brain ischemic stroke: Potential roles of Akt activation and HO-1 induction. Toxicol Appl Pharmacol 2015; 289:142-54. [PMID: 26440581 DOI: 10.1016/j.taap.2015.10.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/22/2015] [Accepted: 10/02/2015] [Indexed: 11/24/2022]
Abstract
The natural product totarol, a phenolic diterpenoid and a major constituent isolated from the sap of Podocarpus totara, has been reported to have a potent antimicrobial activity. In this study, we determined whether totarol possessed an additional neuroprotective activity in vitro and in vivo. We found that totarol prevented glutamate- and oxygen and glucose deprivation-induced neuronal death in primary rat cerebellar granule neuronal cells and cerebral cortical neurons. Totarol increased Akt and GSK-3β phosphorylation, Nrf2 and heme oxygenase-1 (HO-1) protein expressions and suppressed oxidative stress by increasing GSH and SOD activities. The PI3K/Akt inhibitor LY294002 prevented totarol neuroprotective effect by suppressing the totarol-induced changes in HO-1 expression and the activities of GSH and SOD. The HO-1 inhibitor ZnPPIX also prevented totarol-increased GSH and SOD activities. In a model of acute cerebral ischemic injury in Sprague-Dawley rats, produced by occlusion of the middle cerebral artery for 2h followed by 22 h or 46 h of reperfusion, totarol significantly reduced infarct volume and improved the neurological deficit. In this model, totarol increased HO-1 expression and the activities of GSH and SOD. These observations suggest that totarol may be a novel activator of the Akt/HO-1 pathway protecting against ischemic stroke through reduction of oxidative stress.
Collapse
Affiliation(s)
- Yuanxue Gao
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiaojun Xu
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Sai Chang
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yunjie Wang
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yazhou Xu
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Siqi Ran
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Zhangjian Huang
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, PR China
| | - Ping Li
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Road, Shanghai 201203, PR China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Juan M Saavedra
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Hong Liao
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Tao Pang
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China; Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
27
|
Hu S, Cui W, Mak S, Xu D, Hu Y, Tang J, Choi C, Lee M, Pang Y, Han Y. Substantial Neuroprotective and Neurite Outgrowth-Promoting Activities by Bis(propyl)-cognitin via the Activation of Alpha7-nAChR, a Promising Anti-Alzheimer's Dimer. ACS Chem Neurosci 2015; 6:1536-45. [PMID: 26147504 DOI: 10.1021/acschemneuro.5b00108] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The cause of Alzheimer's disease (AD) could be ascribed to the progressive loss of functional neurons in the brain, and hence, agents with neuroprotection and neurite outgrowth-promoting activities that allow for the replacement of lost neurons may have significant therapeutic value. In the current study, the neuroprotective and the neurite outgrowth-promoting activities and molecular mechanisms of bis(propyl)-cognitin (B3C), a multifunctional anti-AD dimer, were investigated. Briefly, B3C (24 h pretreatment) fully protected against glutamate-induced neuronal death in primary cerebellar granule neurons with an IC50 value of 0.08 μM. The neuroprotection of B3C could be abrogated by methyllycaconitine, a specific antagonist of alpha7-nicotinic acetylcholine receptor (α7-nAChR). In addition, B3C significantly promoted neurite outgrowth in both PC12 cells and primary cortical neurons, as evidenced by the increase in the percentage of cells with extended neurites as well as the up-regulation of neuronal markers growth-associated protein-43 and β-III-tubulin. Furthermore, B3C rapidly upregulated the phosphorylation of extracellular signal-regulated kinase (ERK), a critical signaling molecule in neurite outgrowth that is downstream of the α7-nAChR signal pathway. Specific inhibitors of ERK and α7-nAChR, but not those of p38 mitogen-activated protein kinase and c-Jun NH(2)-terminal kinase, blocked the neurite outgrowth as well as ERK activation in PC12 cells induced by B3C. Most importantly, genetic depletion of α7-nAChR significantly abolished B3C-induced neurite outgrowth in PC12 cells. Taken together, our results suggest that B3C provided neuroprotection and neurite outgrowth-promoting activities through the activation of α7-nAChR, which offers a novel molecular insight into the potential application of B3C in AD treatment.
Collapse
Affiliation(s)
- Shengquan Hu
- Department
of Applied Biology and Chemical Technology, Institute of Modern Chinese
Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
- Institute of New Drug Research, Guangdong Province Key Laboratory of Pharmacodynamic, Constituents of Traditional Chinese Medicine & New Drug Research, College of Pharmacy, Jinan University, Guangdong, China
- The Hong Kong Polytechnic University Shenzhen
Research Institute, Shenzhen, China
- State
Key Laboratory of Quality Research in Chinese Medicine, Institute
of Chinese Medical Sciences, University of Macau, Macau, China
| | - Wei Cui
- Department
of Applied Biology and Chemical Technology, Institute of Modern Chinese
Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen
Research Institute, Shenzhen, China
| | - Shinghung Mak
- Department
of Applied Biology and Chemical Technology, Institute of Modern Chinese
Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen
Research Institute, Shenzhen, China
| | - Daping Xu
- Department
of Applied Biology and Chemical Technology, Institute of Modern Chinese
Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen
Research Institute, Shenzhen, China
| | - Yuanjia Hu
- State
Key Laboratory of Quality Research in Chinese Medicine, Institute
of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jing Tang
- Mayo
Cancer Center, Department of Pharmacology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Chunglit Choi
- Department
of Applied Biology and Chemical Technology, Institute of Modern Chinese
Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Mingyuen Lee
- State
Key Laboratory of Quality Research in Chinese Medicine, Institute
of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yuanping Pang
- Mayo
Cancer Center, Department of Pharmacology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Yifan Han
- Department
of Applied Biology and Chemical Technology, Institute of Modern Chinese
Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen
Research Institute, Shenzhen, China
| |
Collapse
|
28
|
Preventing H2O2-induced toxicity in primary cerebellar granule neurons via activating the PI3-K/Akt/GSK3β pathway by kukoamine from Lycii Cortex. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.06.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
29
|
Jiang L, Huang M, Xu S, Wang Y, An P, Feng C, Chen X, Wei X, Han Y, Wang Q. Bis(propyl)-cognitin Prevents β-amyloid-induced Memory Deficits as Well as Synaptic Formation and Plasticity Impairments via the Activation of PI3-K Pathway. Mol Neurobiol 2015; 53:3832-3841. [DOI: 10.1007/s12035-015-9317-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 06/23/2015] [Indexed: 01/09/2023]
|
30
|
Sun Y, Liu WZ, Liu T, Feng X, Yang N, Zhou HF. Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J Recept Signal Transduct Res 2015; 35:600-4. [DOI: 10.3109/10799893.2015.1030412] [Citation(s) in RCA: 902] [Impact Index Per Article: 100.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
31
|
Du WJ, Guo JJ, Gao MT, Hu SQ, Dong XY, Han YF, Liu FF, Jiang S, Sun Y. Brazilin inhibits amyloid β-protein fibrillogenesis, remodels amyloid fibrils and reduces amyloid cytotoxicity. Sci Rep 2015; 5:7992. [PMID: 25613018 PMCID: PMC4303869 DOI: 10.1038/srep07992] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/29/2014] [Indexed: 12/22/2022] Open
Abstract
Soluble amyloid β-protein (Aβ) oligomers, the main neurotoxic species, are predominantly formed from monomers through a fibril-catalyzed secondary nucleation. Herein, we virtually screened an in-house library of natural compounds and discovered brazilin as a dual functional compound in both Aβ42 fibrillogenesis inhibition and mature fibril remodeling, leading to significant reduction in Aβ42 cytotoxicity. The potent inhibitory effect of brazilin was proven by an IC50 of 1.5 ± 0.3 μM, which was smaller than that of (−)-epigallocatechin gallate in Phase III clinical trials and about one order of magnitude smaller than those of curcumin and resveratrol. Most importantly, it was found that brazilin redirected Aβ42 monomers and its mature fibrils into unstructured Aβ aggregates with some β-sheet structures, which could prevent both the primary nucleation and the fibril-catalyzed secondary nucleation. Molecular simulations demonstrated that brazilin inhibited Aβ42 fibrillogenesis by directly binding to Aβ42 species via hydrophobic interactions and hydrogen bonding and remodeled mature fibrils by disrupting the intermolecular salt bridge Asp23-Lys28 via hydrogen bonding. Both experimental and computational studies revealed a different working mechanism of brazilin from that of known inhibitors. These findings indicate that brazilin is of great potential as a neuroprotective and therapeutic agent for Alzheimer's disease.
Collapse
Affiliation(s)
- Wen-Jie Du
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Jing-Jing Guo
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Ming-Tao Gao
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Sheng-Quan Hu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiao-Yan Dong
- 1] Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China [2] Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| | - Yi-Fan Han
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Fu-Feng Liu
- 1] Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China [2] Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| | - Shaoyi Jiang
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195-1750, USA
| | - Yan Sun
- 1] Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China [2] Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| |
Collapse
|
32
|
Hu S, Wang R, Cui W, Zhang Z, Mak S, Xu D, Choi C, Tsim KW, Carlier PR, Lee M, Han Y. Inhibiting β-Amyloid-Associated Alzheimer’s Pathogenesis In Vitro and In Vivo by a Multifunctional Dimeric Bis(12)-hupyridone Derived from Its Natural Analogue. J Mol Neurosci 2014; 55:1014-21. [DOI: 10.1007/s12031-014-0458-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 10/29/2014] [Indexed: 10/24/2022]
|
33
|
Yu H, Yao L, Zhou H, Qu S, Zeng X, Zhou D, Zhou Y, Li X, Liu Z. Neuroprotection against Aβ25–35-induced apoptosis by Salvia miltiorrhiza extract in SH-SY5Y cells. Neurochem Int 2014; 75:89-95. [DOI: 10.1016/j.neuint.2014.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/09/2014] [Accepted: 06/04/2014] [Indexed: 12/30/2022]
|
34
|
MAPK signaling correlates with the antidepressant effects of ketamine. J Psychiatr Res 2014; 55:15-21. [PMID: 24819632 DOI: 10.1016/j.jpsychires.2014.04.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 03/27/2014] [Accepted: 04/10/2014] [Indexed: 11/21/2022]
Abstract
Studies have pointed to a relationship between MAPK kinase (MEK) signaling and the behavioral effects of antidepressant drugs. So, in the present study we examined the behavioral and molecular effects of ketamine, an antagonist of the N-methyl-d-aspartate receptor (NMDA), which has been shown to have an antidepressant effect after the inhibition of MEK signaling in Wistar rats. Our results showed that acute administration of the MEK inhibitor PD184161, produced depressive-like behavior and stopped antidepressant-like effects of ketamine in the forced swimming test. The phosphorylation of extracellular signal-regulated kinase 1/2 (pERK 1/2) was decreased by PD184161 in the amygdala and nucleus accumbens, and the effects of ketamine on pERK 1/2 in the prefrontal cortex and hippocampus were inhibited by PD184161. The ERK 2 levels were decreased by PD184161 in the nucleus accumbens; and the effects of ketamine were blocked in this brain area. The p38 protein kinase (p38MAPK) and proBDNF were inhibited by PD184161, and the MEK inhibitor prevented the effects of ketamine in the nucleus accumbens. In addition, ketamine increased pro-BDNF levels in the hippocampus. In conclusion, our findings demonstrated that an acute blockade of MAPK signaling lead to depressive-like behavior and stopped the antidepressant response of ketamine, suggesting that the effects of ketamine could be mediated, at least in part, by the regulation of MAPK signaling in these specific brain areas.
Collapse
|
35
|
Zhang J, Feng X, Wu J, Xu H, Li G, Zhu D, Yue Q, Liu H, Zhang Y, Sun D, Wang H, Sun J. Neuroprotective effects of resveratrol on damages of mouse cortical neurons induced by β-amyloid through activation of SIRT1/Akt1 pathway. Biofactors 2014; 40:258-67. [PMID: 24132831 DOI: 10.1002/biof.1149] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Revised: 09/04/2013] [Accepted: 09/09/2013] [Indexed: 12/25/2022]
Abstract
Resveratrol (3,5,4'-tihydroxy-trans-stilbene), a polyphenolic phytoalexin found in the skin and seeds of grapes, has been reported to possess a wide range of biological and pharmacological activities including antioxidant, anti-inflammatory, and antimutagenic effects. The present study intended to explore the neuroprotective effects of resveratrol against Aβ25-35 -induced neurotoxicity of cultured mouse cortical neurons and the possible mechanisms involved. For this purpose, mouse cortical neurons were cultured and exposed to 30 μM Aβ25-35 in the absence or presence of resveratrol (5, 10, and 25 μM). In addition, the potential contribution of the SIRT1/Akt1 neuroprotective pathway in resveratrol-mediated protection against Aβ25-35 -induced neurotoxicity was also investigated. The results showed that resveratrol dose-dependently increased cell viability and reduced the number of apoptotic cells as measured by 3-[4,5-dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide (MTT) assay, lactate dehydrogenase (LDH) activity assay, reactive oxygen species (ROS) activity assay, and Hoechst/PI double staining. Further study revealed that resveratrol through activation of SIRT1/Akt1 to avert apoptosis. These findings raise the possibility that resveratrol may be a potent therapeutic compound against the neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Anatomy, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Cacabelos R, Cacabelos P, Torrellas C, Tellado I, Carril JC. Pharmacogenomics of Alzheimer's disease: novel therapeutic strategies for drug development. Methods Mol Biol 2014; 1175:323-556. [PMID: 25150875 DOI: 10.1007/978-1-4939-0956-8_13] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a major problem of health and disability, with a relevant economic impact on our society. Despite important advances in pathogenesis, diagnosis, and treatment, its primary causes still remain elusive, accurate biomarkers are not well characterized, and the available pharmacological treatments are not cost-effective. As a complex disorder, AD is a polygenic and multifactorial clinical entity in which hundreds of defective genes distributed across the human genome may contribute to its pathogenesis. Diverse environmental factors, cerebrovascular dysfunction, and epigenetic phenomena, together with structural and functional genomic dysfunctions, lead to amyloid deposition, neurofibrillary tangle formation, and premature neuronal death, the major neuropathological hallmarks of AD. Future perspectives for the global management of AD predict that genomics and proteomics may help in the search for reliable biomarkers. In practical terms, the therapeutic response to conventional drugs (cholinesterase inhibitors, multifactorial strategies) is genotype-specific. Genomic factors potentially involved in AD pharmacogenomics include at least five categories of gene clusters: (1) genes associated with disease pathogenesis; (2) genes associated with the mechanism of action of drugs; (3) genes associated with drug metabolism (phase I and II reactions); (4) genes associated with drug transporters; and (5) pleiotropic genes involved in multifaceted cascades and metabolic reactions. The implementation of pharmacogenomic strategies will contribute to optimize drug development and therapeutics in AD and related disorders.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Chair of Genomic Medicine, Camilo José Cela University, 28692, Villanueva de la Cañada, Madrid, Spain,
| | | | | | | | | |
Collapse
|
37
|
Wang J, Pang T, Hafko R, Benicky J, Sanchez-Lemus E, Saavedra JM. Telmisartan ameliorates glutamate-induced neurotoxicity: roles of AT(1) receptor blockade and PPARγ activation. Neuropharmacology 2013; 79:249-61. [PMID: 24316465 DOI: 10.1016/j.neuropharm.2013.11.022] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/22/2013] [Accepted: 11/26/2013] [Indexed: 01/22/2023]
Abstract
Sartans (Angiotensin II AT(1) Receptor Blockers, ARBs) are powerful neuroprotective agents in vivo and protect against IL-1β neurotoxicity in vitro. The purpose of this research was to determine the extent of sartan neuroprotection against glutamate excitotoxicity, a common cause of neuronal injury and apoptosis. The results show that sartans are neuroprotective, significantly reducing glutamate-induced neuronal injury and apoptosis in cultured rat primary cerebellar granule cells (CGCs). Telmisartan was the most potent sartan studied, with an order of potency telmisartan > candesartan > losartan > valsartan. Mechanisms involved reduction of pro-apoptotic caspase-3 activation, protection of the survival PI3K/Akt/GSK-3β pathway and prevention of glutamate-induced ERK1/2 activation. NMDA receptor stimulation was essential for glutamate-induced cell injury and apoptosis. Participation of AT(1A) receptor was supported by glutamate-induced upregulation of AT(1A) gene expression and AT(1) receptor binding. Conversely, AT(1B) or AT(2) receptors played no role. Glutamate-induced neuronal injury and the neuroprotective effect of telmisartan were decreased, but not abolished, in CGCs obtained from AT(1A) knock-out mice. This indicates that although AT(1) receptors are necessary for glutamate to exert its full neurotoxic potential, part of the neuroprotective effect of telmisartan is independent of AT(1) receptor blockade. PPARγ activation was also involved in the neuroprotective effects of telmisartan, as telmisartan enhanced PPARγ nuclear translocation and the PPARγ antagonist GW9662 partially reversed the neuroprotective effects of telmisartan. The present results substantiate the therapeutic use of sartans, in particular telmisartan, in neurodegenerative diseases and traumatic brain disorders where glutamate neurotoxicity plays a significant role.
Collapse
Affiliation(s)
- Juan Wang
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA.
| | - Tao Pang
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA; New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, PR China
| | - Roman Hafko
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA
| | - Julius Benicky
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA.
| | - Enrique Sanchez-Lemus
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA.
| | - Juan M Saavedra
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA; Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
38
|
Hu SQ, Cui W, Xu DP, Mak SH, Tang J, Choi CL, Pang YP, Han YF. Substantial neuroprotection against K+ deprivation-induced apoptosis in primary cerebellar granule neurons by novel dimer bis(propyl)-cognitin via the activation of VEGFR-2 signaling pathway. CNS Neurosci Ther 2013; 19:764-72. [PMID: 23826635 DOI: 10.1111/cns.12141] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 05/09/2013] [Accepted: 05/10/2013] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Neuronal loss via apoptosis in CNS is the fundamental mechanism underlying various neurodegenerative diseases. Compounds with antiapoptotic property might have therapeutic effects for these diseases. In this study, bis(propyl)-cognitin (B3C), a novel dimer that possesses anti-AChE and anti-N-methyl-d-aspartate receptor activities, was investigated for its neuroprotective effect on K(+) deprivation-induced apoptosis in cerebellar granule neurons (CGNs). METHODS Cerebellar granule neurons were switched to K(+) deprived medium with or without B3C. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium assay, fluorescein diacetate (FDA)/propidium iodide (PI) staining, Hoechst staining, and DNA laddering assays were applied to detect cytotoxicity and apoptosis. Additionally, the expression of p-VEGFR-2, p-Akt, p-glycogen synthase kinase 3β (GSK3β), and p-extracellular signal-regulated kinase (ERK) was examined in CGNs. RESULTS Switching CGNs to K(+) deprived medium resulted in remarkable apoptosis, which could be substantially blocked by B3C treatment (IC50 , 0.37 μM). Moreover, a rapid decrease in p-Tyr1054-VEGFR-2 was observed after the switch. B3C significantly reversed the inhibition of p-Tyr1054-VEGFR-2 as well as Akt and ERK pathways. VEGFR-2 inhibitor PTK787/ZK222584, as well as PI3-K inhibitor LY294002 and MEK inhibitor PD98059, each abolished the neuroprotective effect of B3C. CONCLUSIONS Our results demonstrate that B3C blocks K(+) deprivation-induced apoptosis in CGNs through regulating VEGFR-2/Akt/GSK3β and VEGFR-2/ERK signaling pathways, providing a molecular insight into the therapeutic potential of B3C for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sheng-Quan Hu
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; The Hong Kong Polytechnic University, Shenzhen Research Institute, Shenzhen, China
| | | | | | | | | | | | | | | |
Collapse
|