1
|
Che X, Zhao Y, Xu Z, Hu Y, Ren A, Wu C, Yang J. Unlocking the Potential of l-α-Glycerylphosphorylcholine: From Metabolic Pathways to Therapeutic Applications. Nutr Rev 2025:nuaf008. [PMID: 40036805 DOI: 10.1093/nutrit/nuaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025] Open
Abstract
l-α-Glycerylphosphorylcholine (GPC), also known as choline alphoscerate or α-glycerophosphorylcholine, serves as both a pharmaceutical product and a dietary supplement. Through its metabolic pathways, GPC acts as the precursor not only of choline and acetylcholine but also of various phospholipids. Extensive preclinical and clinical evidence demonstrates that GPC effectively alleviates cognitive impairment associated with Alzheimer's disease, vascular dementia, cerebral ischemia, stress, and epilepsy, among other conditions. Additionally, GPC has beneficial effects on such conditions and measures as ischemic/hypoxic conditions, ionizing radiation-induced damage, exercise performance, growth hormone release, and liver damage. As well as facilitating cholinergic neurotransmission, evidence also indicates GPC, among other activities, also can promote γ-aminobutyric acid release, enhance protein kinase C activity, facilitate hippocampal neurogenesis, upregulate neurotrophic factors, and inhibit inflammation. In preclinical studies, results indicate that GPC is not genotoxic in vitro or in vivo. Extensive human studies indicate GPC causes no severe adverse effects. Possible risks of atherosclerosis and stroke await necessary validation. In this review, the GPC-related metabolic pathways, pharmacological effects, mechanisms of action, and safety evaluation are discussed with the aim of providing a comprehensive understanding of GPC.
Collapse
Affiliation(s)
- Xiaohang Che
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yang Zhao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhongtian Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yue Hu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Aoxin Ren
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
2
|
Reda SM, Setti SE, Berthiaume AA, Wu W, Taylor RW, Johnston JL, Stein LR, Moebius HJ, Church KJ. Fosgonimeton attenuates amyloid-beta toxicity in preclinical models of Alzheimer's disease. Neurotherapeutics 2024; 21:e00350. [PMID: 38599894 PMCID: PMC11067346 DOI: 10.1016/j.neurot.2024.e00350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/13/2024] [Accepted: 03/16/2024] [Indexed: 04/12/2024] Open
Abstract
Positive modulation of hepatocyte growth factor (HGF) signaling may represent a promising therapeutic strategy for Alzheimer's disease (AD) based on its multimodal neurotrophic, neuroprotective, and anti-inflammatory effects addressing the complex pathophysiology of neurodegeneration. Fosgonimeton is a small-molecule positive modulator of the HGF system that has demonstrated neurotrophic and pro-cognitive effects in preclinical models of dementia. Herein, we evaluate the neuroprotective potential of fosgonimeton, or its active metabolite, fosgo-AM, in amyloid-beta (Aβ)-driven preclinical models of AD, providing mechanistic insight into its mode of action. In primary rat cortical neurons challenged with Aβ (Aβ1-42), fosgo-AM treatment significantly improved neuronal survival, protected neurite networks, and reduced tau hyperphosphorylation. Interrogation of intracellular events indicated that cortical neurons treated with fosgo-AM exhibited a significant decrease in mitochondrial oxidative stress and cytochrome c release. Following Aβ injury, fosgo-AM significantly enhanced activation of pro-survival effectors ERK and AKT, and reduced activity of GSK3β, one of the main kinases involved in tau hyperphosphorylation. Fosgo-AM also mitigated Aβ-induced deficits in Unc-like kinase 1 (ULK1) and Beclin-1, suggesting a potential effect on autophagy. Treatment with fosgo-AM protected cortical neurons from glutamate excitotoxicity, and such effects were abolished in the presence of an AKT or MEK/ERK inhibitor. In vivo, fosgonimeton administration led to functional improvement in an intracerebroventricular Aβ25-35 rat model of AD, as it significantly rescued cognitive function in the passive avoidance test. Together, our data demonstrate the ability of fosgonimeton to counteract mechanisms of Aβ-induced toxicity. Fosgonimeton is currently in clinical trials for mild-to-moderate AD (NCT04488419; NCT04886063).
Collapse
Affiliation(s)
- Sherif M Reda
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Sharay E Setti
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | | | - Wei Wu
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Robert W Taylor
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Jewel L Johnston
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Liana R Stein
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Hans J Moebius
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA
| | - Kevin J Church
- Athira Pharma, Inc., 18706 North Creek Parkway, Suite 104, Bothell, WA, 98011, USA.
| |
Collapse
|
3
|
Lee SY, Cho HY, Oh JP, Park J, Bae SH, Park H, Kim EJ, Lee JH. Therapeutic Effects of Combination of Nebivolol and Donepezil: Targeting Multifactorial Mechanisms in ALS. Neurotherapeutics 2023; 20:1779-1795. [PMID: 37782409 PMCID: PMC10684847 DOI: 10.1007/s13311-023-01444-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2023] [Indexed: 10/03/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by progressive loss of motor neurons in the spinal cord. Although the disease's pathophysiological mechanism remains poorly understood, multifactorial mechanisms affecting motor neuron loss converge to worsen the disease. Although two FDA-approved drugs, riluzole and edaravone, targeting excitotoxicity and oxidative stress, respectively, are available, their efficacies are limited to extending survival by only a few months. Here, we developed combinatorial drugs targeting multifactorial mechanisms underlying key components in ALS disease progression. Using data analysis based on the genetic information of patients with ALS-derived cells and pharmacogenomic data of the drugs, a combination of nebivolol and donepezil (nebivolol-donepezil) was identified for ALS therapy. Here, nebivolol-donepezil markedly reduced the levels of cytokines in the microglial cell line, inhibited nuclear factor-κB (NF-κB) nucleus translocation in the HeLa cell and substantially protected against excitotoxicity-induced neuronal loss by regulating the PI3K-Akt pathway. Nebivolol-donepezil significantly promoted the differentiation of neural progenitor cells (NPC) into motor neurons. Furthermore, we verified the low dose efficacy of nebivolol-donepezil on multiple indices corresponding to the quality of life of patients with ALS in vivo using SOD1G93A mice. Nebivolol-donepezil delayed motor function deterioration and halted motor neuronal loss in the spinal cord. Drug administration effectively suppressed muscle atrophy by mitigating the proportion of smaller myofibers and substantially reducing phospho-neurofilament heavy chain (pNF-H) levels in the serum, a promising ALS biomarker. High-dose nebivolol-donepezil significantly prolonged survival and delayed disease onset compared with vehicle-treated mice. These results indicate that the combination of nebivolol-donepezil efficiently prevents ALS disease progression, benefiting the patients' quality of life and life expectancy.
Collapse
Affiliation(s)
- Soo Yeon Lee
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Hye-Yeon Cho
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Jung-Pyo Oh
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Jiae Park
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Sang-Hun Bae
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Haesun Park
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Eun Jung Kim
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea.
| | - Ji-Hyun Lee
- DR. NOAH BIOTECH Inc., 91, Changnyong-daero 256beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea.
| |
Collapse
|
4
|
Ameliorative effects of bromelain on aluminum-induced Alzheimer's disease in rats through modulation of TXNIP pathway. Int J Biol Macromol 2023; 227:1119-1131. [PMID: 36462588 DOI: 10.1016/j.ijbiomac.2022.11.291] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/01/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
Alzheimer's disease (AD) is known as "type 3 diabetes". As thioredoxin binding protein (TXNIP) has been shown to be involved in brain insulin resistance, the present study evaluated the roles of TXNIP, phospho-insulin receptor substrate 1 (P-IRS-1), and phosphatidyl inositol-3 kinase (PI3K) in the pathogenesis of AD. The potential ameliorative effect of bromelain compared to donepezil was evaluated in an aluminum chloride (AlCl3)-induced AD in rats. Behavioral tests demonstrated similar improvements in exploratory activity, cognitive and spatial memory functions, anxiety, and depression levels between rats treated with bromelain and donepezil. Donepezil was superior to bromelain in improving locomotor activity. Histopathological examinations demonstrated neuronal degeneration in the AlCl3 group that was almost normalized by bromelain and donepezil. Moreover, there was deposition of amyloid plaques in the AlCl3 group that was improved by bromelain and donepezil. Acetylcholine esterase levels were significantly increased in rats treated with AlCl3 group and significantly decreased in rats treated with bromelain and donepezil. Furthermore, AlCl3 group showed a significantly increased TXNIP and P-IRS1 and a significantly reduced PI3K levels. These effects were ameliorated by bromelain and donepezil treatment. The present study demonstrates a previously unreported modulatory effect of bromelain on the TXNIP/P-IRS-1/PI3K axis in AD model.
Collapse
|
5
|
Bhat JA, Kumar M. Neuroprotective Effects of Theobromine in permanent bilateral common carotid artery occlusion rat model of cerebral hypoperfusion. Metab Brain Dis 2022; 37:1787-1801. [PMID: 35587851 DOI: 10.1007/s11011-022-00995-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
Abstract
Cerebral hypoperfusion (CH) is a common underlying mechanism of dementia disorders linked to aberrations in the neurovascular unit. Hemodynamic disturbances adversely affect cellular energy homeostasis that triggers a sequence of events leading to irrevocable damage to the brain and neurobehavioral discrepancies. Theobromine is a common ingredient of many natural foods consumed by a large population worldwide. Theobromine has shown health benefits in several studies, attributed to regulation of calcium homeostasis, phosphodiesterase, neurotransmission, and neurotrophins. The current study evaluated the neuroprotective potential of theobromine against CH in the permanent bilateral common carotid artery occlusion (BCCAO) prototype. Wistar rats were distributed in Sham-operated (S), S + T100, CH, CH + T50, and CH + T100 groups. Animals received permanent BCCAO or Sham treatment on day 1. Theobromine (50, 100 mg/kg) was given orally in animals subjected to BCCAO for 14 days daily. CH caused neurological deficits (12-point scale), motor dysfunction, and memory impairment in rats. Treatment with theobromine significantly attenuated neurological deficits and improved sensorimotor functions and memory in rats with CH. In biochemistry investigation of the entire brain, findings disclosed reduction in brain oxidative stress, inflammatory intermediaries (tumor necrosis factor-α, interleukin-1β and - 6, nuclear factor-κB), markers of cell demise (lactate dehydrogenase, caspase-3), acetylcholinesterase activity, and improvement in γ-aminobutyric acid quantity in rats that were given theobromine for 14 days daily after CH. Histopathological analysis substantiated attenuation of neurodegenerative changes by theobromine. The findings of this study indicated that theobromine could improve neurological scores, sensorimotor abilities, and memory in CH prototype.
Collapse
Affiliation(s)
- Javeed Ahmad Bhat
- Department of Pharmacology, Swift School of Pharmacy, Ghaggar Sarai, Rajpura, Punjab, India
| | - Manish Kumar
- Department of Pharmacology, Swift School of Pharmacy, Ghaggar Sarai, Rajpura, Punjab, India.
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| |
Collapse
|
6
|
An T, Lu Y, Yan X, Hou J. Insights Into the Properties, Biological Functions, and Regulation of USP21. Front Pharmacol 2022; 13:944089. [PMID: 35846989 PMCID: PMC9279671 DOI: 10.3389/fphar.2022.944089] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/14/2022] [Indexed: 11/20/2022] Open
Abstract
Deubiquitylating enzymes (DUBs) antagonize ubiquitination by removing ubiquitin from their substrates. The role of DUBs in controlling various physiological and pathological processes has been extensively studied, and some members of DUBs have been identified as potential therapeutic targets in diseases ranging from tumors to neurodegeneration. Ubiquitin-specific protease 21 (USP21) is a member of the ubiquitin-specific protease family, the largest subfamily of DUBs. Although USP21 was discovered late and early research progress was slow, numerous studies in the last decade have gradually revealed the importance of USP21 in a wide variety of biological processes. In particular, the pro-carcinogenic effect of USP21 has been well elucidated in the last 2 years. In the present review, we provide a comprehensive overview of the current knowledge on USP21, including its properties, biological functions, pathophysiological roles, and cellular regulation. Limited pharmacological interventions for USP21 have also been introduced, highlighting the importance of developing novel and specific inhibitors targeting USP21.
Collapse
Affiliation(s)
- Tao An
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Yanting Lu
- College of TCM, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xu Yan
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Jingjing Hou
- Department of Gastrointestinal Surgery, School of Medicine, Institute of Gastrointestinal Oncology, Zhongshan Hospital of Xiamen University, Xiamen University, Xiamen, China
- *Correspondence: Jingjing Hou,
| |
Collapse
|
7
|
Remya C, Dileep KV, Variyar EJ, Zhang KYJ, Omkumar RV, Sadasivan C. Chemical similarity assisted search for acetylcholinesterase inhibitors: Molecular modeling and evaluation of their neuroprotective properties. Int J Biol Macromol 2021; 174:466-476. [PMID: 33497692 DOI: 10.1016/j.ijbiomac.2021.01.148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/07/2021] [Accepted: 01/20/2021] [Indexed: 01/02/2023]
Abstract
Alzheimer's disease (AD) is an obstinate and progressive neurodegenerative disorder, mainly characterized by cognitive decline. Increasing number of AD patients and the lack of promising treatment strategies demands novel therapeutic agents to combat various disease pathologies in AD. Recent progresses in understanding molecular mechanisms in AD helped researchers to streamline the various therapeutic approaches. Inhibiting acetylcholinesterase (AChE) activity has emerged as one of the potential treatment strategies. The present study discusses the identification of two potent AChE inhibitors (ZINC11709541 and ZINC11996936) from ZINC database through conventional in silico approaches and their in vitro validations. These inhibitors have strong preferences towards AChE than butyrylcholinesterase (BChE) and didn't evoke any significant reduction in the cell viability of HEK-293 cells and primary cortical neurons. Furthermore, promising neuroprotective properties has also been displayed against glutamate induced excitotoxicity in primary cortical neurons. The present study proposes two potential drug lead compounds for the treatment of AD, that can be used for further studies and preclinical evaluation.
Collapse
Affiliation(s)
- Chandran Remya
- Department of Biotechnology and Microbiology, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala 670661, India
| | - K V Dileep
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - E Jayadevi Variyar
- Department of Biotechnology and Microbiology, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala 670661, India
| | - Kam Y J Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - R V Omkumar
- Molecular Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud P. O., Trivandrum, Kerala 695014, India
| | - C Sadasivan
- Department of Biotechnology and Microbiology, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala 670661, India.
| |
Collapse
|
8
|
Sun RX, Sun ZH, Ren Q, Li L, Yin L, Li F, Su X. Gadd45α affects retinal ganglion cell injury in chronic ocular hypertension rats by regulating p38MAPK pathway. Gene 2020; 763:145030. [PMID: 32755658 DOI: 10.1016/j.gene.2020.145030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the impact and the mechanism of Gadd45α mediating p38MAPK pathway on the retinal ganglion cells (RGCs) injury in chronic ocular hypertension (COH) rats. METHODS COH model in rats were established and intraocular pressure (IOP) was tested. Retrograde labeling was used for counting RGCs and TUNEL staining was performed for RGCs apoptosis. Western Blotting was conducted to examine the expression of Gadd45α and p38MAPK pathway. Besides, RGC-5 cells cultured in vitro were treated with H2O2. Cell viability was detected by CCK-8, ROS level tested by DCFH-DA assay, and cell apoptosis examined by flow cytometry. RESULTS COH rats had increased expression of Gadd45α and p-p38/p38 protein 1-4 weeks after surgery. Rats in COH group enhanced obviously in IOP, RGC apoptosis rate and the protein expression of Gadd45α, p-p38/p38, Bax/Bcl-2 and cleaved caspase-3, but declined appreciably in RGC counting. However, the above indicators of COH rats were effectively improved by Gadd45α shRNA treatment. Additionally, RGC-5 cells in H2O2 group reduced in cell viability and went up in ROS level and apoptosis rate. The H2O2-induced RGC-5 cells treated with Gadd45α shRNA were improved apparently in those indicators, and cells treated with pcDNA Gadd45α showed an opposite trend. Moreover, p38 MAPK inhibitor SB203580 can effectively reverse the damage of pcDNA Gadd45α from H2O2-induced RGC-5 cells. CONCLUSION Silencing Gadd45α can reduce the RGC damage in COH rats by inhibiting p38MAPK pathway and such a protective role may be associated with the suppression of RGC apoptosis and the mitigation of oxidative stress.
Collapse
Affiliation(s)
- Rui-Xue Sun
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, Shijiazhuang 050000, Hebei Province, China
| | - Zhao-Hui Sun
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, Shijiazhuang 050000, Hebei Province, China
| | - Qian Ren
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, Shijiazhuang 050000, Hebei Province, China
| | - Li Li
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, Shijiazhuang 050000, Hebei Province, China
| | - Li Yin
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, Shijiazhuang 050000, Hebei Province, China
| | - Fang Li
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, Shijiazhuang 050000, Hebei Province, China
| | - Xian Su
- Department of Ophthalmology, The First Hospital of Shijiazhuang City, Shijiazhuang 050000, Hebei Province, China.
| |
Collapse
|
9
|
Zaitone SA, Alshaman R, Alattar A, Elsherbiny NM, Abogresha NM, El-Kherbetawy MK, Elaskary AA, Hashish AA, Rashed LA, Ahmed E. Retinoprotective effect of donepezil in diabetic mice involves mitigation of excitotoxicity and activation of PI3K/mTOR/BCl 2 pathway. Life Sci 2020; 262:118467. [PMID: 32961236 DOI: 10.1016/j.lfs.2020.118467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022]
Abstract
Donepezil (DNPZ) has shown neuroprotective effect in many disorders. The current study tested the putative retinoprotection provided by donepezil in mouse diabetic retinopathy. Swiss albino mice were allocated to, 1] saline control, 2] diabetic, 3&4] diabetic+DNPZ (1 or 4 mg/kg). After induction of diabetes, mice were maintained for 8 weeks then DNPZ therapy was launched for 28 days. Retinas were isolated and used for histopathology and immunohistochemistry for caspase 3 and the anti-apoptotic protein, B-cell lymphoma 2 (BCl2). Retinas were examined for glutamate, acetylcholine and oxidation markers. Western blot analysis measured inflammatory cytokines, N-methyl-d-aspartate receptors (NMDARs), phosphorylated and total phosphatidylinositol-3 kinase and mTOR, BCl2 and cleaved caspase 3. Significant histopathological changes and decreased thickness were found in diabetic retinas (125.52 ± 2.85 vs. 157.15 ± 7.55 in the saline group). In addition, retinal glutamate (2.39-fold), inflammatory cytokines and NMDARs proteins (4.9-fold) were higher in the diabetic retinas. Western blot analysis revealed low ratio of phosphorylated/total PI3K (0.21 ± 0.043 vs. 1 ± 0.005) and mTOR (0.18 ± 0.04 vs. 1 ± 0.005), low BCl2 (0.28 ± 0.06 vs. 1 ± 0.005) and upregulated cleaved caspase 3 (5.18 ± 1.27 vs. 1 ± 0.05 in the saline group) versus the saline control. DNPZ ameliorated the histopathologic manifestations and to prevent the decrease in retinal thickness. DNPZ (4 mg/kg) improved phosphorylation of PI3K (0.76 ± 0.12 vs. 0.21 ± 0.04) and mTOR (0.59 ± 0.09 vs. 0.18 ± 0.04) and increased BCl2 (0.75 ± 0.08 vs. 0.28 ± 0.06) versus the diabetic control group. This study explained the retinoprotective effect of DNPZ in mouse diabetic retinopathy and highlighted that mitigation of excitotoxicity, improving phosphorylation of PI3K/mTOR and increasing BCl2 contribute to this effect.
Collapse
Affiliation(s)
- Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, 71491 Tabuk, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt.
| | - Reem Alshaman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, 71491 Tabuk, Saudi Arabia
| | - Abdullah Alattar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, 71491 Tabuk, Saudi Arabia
| | - Nehal M Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, 71491 Tabuk, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Noha M Abogresha
- Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | | | - Abdullah A Hashish
- Clinical Pathology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Laila A Rashed
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Eman Ahmed
- Clinical Pharmacology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
10
|
Cheng Z, Zhang T, Zheng J, Ding W, Wang Y, Li Y, Zhu L, Murray M, Zhou F. Betulinic acid derivatives can protect human Müller cells from glutamate-induced oxidative stress. Exp Cell Res 2019; 383:111509. [PMID: 31344390 DOI: 10.1016/j.yexcr.2019.111509] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 02/06/2023]
Abstract
Müller cells are the predominant retinal glial cells. One of the key roles of Müller cells is in the uptake of the neurotransmitter glutamate and in its conversion to glutamine. Müller cell dysfunction due to oxidative stress elicited by high glutamate concentrations can lead to toxicity, which promote the pathogenesis of retinal diseases like diabetic retinopathy and glaucoma. This study investigated the anti-oxidant activity and mechanisms of betulinic acid (BA) and its derivatives in human Müller cells. Human MIO-M1 Müller cells were pre-treated in the presence or absence of BA, BE as well as their derivatives (named H3-H20) followed by incubation with glutamate. Cell viability was evaluated with the MTT and calcein-AM assays. Reactive oxygen species (ROS) production in MIO-M1 cells was measured using CM-H2DCFDA and flow cytometry. The activation of cellular apoptosis and necrosis was analyzed with annexin V/PI staining and flow cytometry. The modulation of signaling pathways involved in glutamate-mediated cytotoxicity and ROS production was evaluated by immunoblotting. The BA derivatives H3, H5 and H7 exhibited minimal cytotoxicity and significant anti-oxidant activity. These compounds significantly suppressed ROS production and attenuated cellular necrosis elicited by glutamate-induced oxidative stress. The protective effects of H3, H5 and H7 in MIO-M1 cells were associated with the attenuation of Akt, Erk, and JNK signaling. The BA analogues H3, H5 and H7 are protective against glutamate-induced oxidative stress in human Müller cells, and elicit their actions by modulation of the Erk, Akt and JNK signaling pathways. These agents are potential candidate molecules for the prevention or treatment of human retinal diseases.
Collapse
Affiliation(s)
- Zhengqi Cheng
- The University of Sydney, School of Pharmacy, NSW, 2006, Australia
| | - Ting Zhang
- The University of Sydney, Save Sight Institute, Sydney, NSW, 2000, Australia
| | - Jian Zheng
- Northeast Forestry University, Center for Bioactive Products/Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, 150040, China
| | - Weimin Ding
- Harbin University of Science and Technology, School of Chemical and Environmental Engineering, Harbin, 150080, Heilongjiang, China
| | - Yang Wang
- Northeast Forestry University, Center for Bioactive Products/Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, 150040, China
| | - Yue Li
- The University of Sydney, School of Pharmacy, NSW, 2006, Australia
| | - Ling Zhu
- The University of Sydney, Save Sight Institute, Sydney, NSW, 2000, Australia
| | - Michael Murray
- The University of Sydney, Discipline of Pharmacology, Faculty of Medicine and Health, NSW, 2006, Australia
| | - Fanfan Zhou
- The University of Sydney, School of Pharmacy, NSW, 2006, Australia.
| |
Collapse
|
11
|
Atef MM, El-Sayed NM, Ahmed AAM, Mostafa YM. Donepezil improves neuropathy through activation of AMPK signalling pathway in streptozotocin-induced diabetic mice. Biochem Pharmacol 2018; 159:1-10. [PMID: 30414938 DOI: 10.1016/j.bcp.2018.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/07/2018] [Indexed: 12/16/2022]
Abstract
Diabetic neuropathy (DN) is a common complication of diabetes mellitus and is associated with structural changes in the nerves. However, the molecular basis for DN is poorly understood. Adenosine monophosphate activated protein kinase (AMPK) has been shown to regulate the activity of some kinases including protein kinase B (AKT), mitogen-activated protein kinases (MAPK) and mammalian target of rapamycin complex 1 (mTORC1) that represent important signalling pathways modulating the function of peripheral nociceptive neuron. Donepezil can activate AMPK and exerts neuroprotective effects. In this study, streptozotocin (45 mg/kg for 5 Day, i.p.) was used to induce experimental DN. After confirmation of development of neuropathy, mice were randomly distributed into five groups: Group 1; negative control group received saline (0.9%NaCl), Group 2; diabetic mice received saline, Group (3-5); diabetic mice received daily donepezil (1, 2 or 4 mg/kg, p.o.) respectively for 20 days. Mice were then sacrificed under anesthesia then their sciatic nerve and spinal cord were dissected out and processed for biochemical and histopathological studies. Diabetic mice revealed severe histological abnormalities including degenerated neurons in the spinal cord and swollen myelin sheath with inflammatory edema observed in sciatic nerves. In addition, diabetic mice showed reduced expression of p-AMPK in sciatic nerves with consequent activation of AKT/MAPK/4EBP1. A significant upregulation of the N-Methyl-d-aspartate (NMDA) receptors in both cervical and lumbar regions of spinal cord of diabetic mice was also demonstrated. Donepezil, an AMPK activator, blocked the phosphorylation of AKT/MAPK/4EBP1, down regulate the expression of NMDA receptors and reversed hyperalgesia developed in diabetic mice. Therefore, Donepezil could be a potential pharmacological agent for management of DN.
Collapse
Affiliation(s)
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt.
| | - Amal A M Ahmed
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Yasser M Mostafa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
12
|
Haraguchi Y, Mizoguchi Y, Ohgidani M, Imamura Y, Murakawa-Hirachi T, Nabeta H, Tateishi H, Kato TA, Monji A. Donepezil suppresses intracellular Ca 2+ mobilization through the PI3K pathway in rodent microglia. J Neuroinflammation 2017; 14:258. [PMID: 29273047 PMCID: PMC5741946 DOI: 10.1186/s12974-017-1033-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022] Open
Abstract
Background Microglia are resident innate immune cells which release many factors including proinflammatory cytokines or nitric oxide (NO) when they are activated in response to immunological stimuli. Pathophysiology of Alzheimer’s disease (AD) is related to the inflammatory responses mediated by microglia. Intracellular Ca2+ signaling is important for microglial functions such as release of NO and cytokines. In addition, alteration of intracellular Ca2+ signaling underlies the pathophysiology of AD, while it remains unclear how donepezil, an acetylcholinesterase inhibitor, affects intracellular Ca2+ mobilization in microglial cells. Methods We examined whether pretreatment with donepezil affects the intracellular Ca2+ mobilization using fura-2 imaging and tested the effects of donepezil on phagocytic activity by phagocytosis assay in rodent microglial cells. Results In this study, we observed that pretreatment with donepezil suppressed the TNFα-induced sustained intracellular Ca2+ elevation in both rat HAPI and mouse primary microglial cells. On the other hand, pretreatment with donepezil did not suppress the mRNA expression of both TNFR1 and TNFR2 in rodent microglia we used. Pretreatment with acetylcholine but not donepezil suppressed the TNFα-induced intracellular Ca2+ elevation through the nicotinic α7 receptors. In addition, sigma 1 receptors were not involved in the donepezil-induced suppression of the TNFα-mediated intracellular Ca2+ elevation. Pretreatment with donepezil suppressed the TNFα-induced intracellular Ca2+ elevation through the PI3K pathway in rodent microglial cells. Using DAF-2 imaging, we also found that pretreatment with donepezil suppressed the production of NO induced by TNFα treatment and the PI3K pathway could be important for the donepezil-induced suppression of NO production in rodent microglial cells. Finally, phagocytosis assay showed that pretreatment with donepezil promoted phagocytic activity of rodent microglial cells through the PI3K but not MAPK/ERK pathway. Conclusions These suggest that donepezil could directly modulate the microglial function through the PI3K pathway in the rodent brain, which might be important to understand the effect of donepezil in the brain. Electronic supplementary material The online version of this article (10.1186/s12974-017-1033-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yoshinori Haraguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Yoshito Mizoguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| | - Masahiro Ohgidani
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshiomi Imamura
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Toru Murakawa-Hirachi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hiromi Nabeta
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hiroshi Tateishi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akira Monji
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| |
Collapse
|
13
|
Zhou X, Cheng Y, Zhang R, Li G, Yang B, Zhang S, Wu J. Alpha7 nicotinic acetylcholine receptor agonist promotes retinal ganglion cell function via modulating GABAergic presynaptic activity in a chronic glaucomatous model. Sci Rep 2017; 7:1734. [PMID: 28496108 PMCID: PMC5431927 DOI: 10.1038/s41598-017-02092-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/05/2017] [Indexed: 01/01/2023] Open
Abstract
Alpha-7 nicotinic acetylcholine receptor (α7-nAChR) agonists can prevent glutamate-induced excitotoxicity in cultured retinal ganglion cells (RGCs). However, the neuroprotective effects and the mechanism of action of PNU-282987, an α7-nAChR agonist, in a chronic in vivo rat glaucoma model are poorly understood. We found that elevated intraocular pressure (IOP) downregulated retinal α7-nAChR expression. Electroretinography revealed that the amplitude of the photopic negative response (PhNR) decreased in parallel with the loss of RGCs caused by elevated IOP. PNU-282987 enhanced RGC viability and function and decreased terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive signals in RGCs. Patch-clamp recordings revealed differences in the baseline frequencies and decay times of the miniature GABAergic inhibitory postsynaptic currents (mIPSCs) of RGCs between control and glaucomatous retinal slices. The results of western blotting and immunostaining showed that glutamic acid decarboxylase 65/67 and GABA deficits persisted in glaucomatous retinas and that these deficits were reversed by PNU-282987. Patch-clamp recordings also showed that PNU-282987 significantly increased the frequency and amplitude of the GABAergic mIPSCs of RGCs. The protective effects of PNU-292987 were blocked by intravitreal administration of selective GABAA receptor antagonists. The modulation of GABAergic synaptic transmission by PNU-282987 causes de-excitation of ganglion cell circuits and suppresses excitotoxic processes.
Collapse
Affiliation(s)
- Xujiao Zhou
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, 200032, China.,Key Laboratory of Myopia, Ministry of Health, Shanghai, 200032, China
| | - Yun Cheng
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rong Zhang
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Gang Li
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Boqi Yang
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shenghai Zhang
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jihong Wu
- Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, 200032, China. .,Key Laboratory of Myopia, Ministry of Health, Shanghai, 200032, China.
| |
Collapse
|
14
|
Amani N, Soodi M, Daraei B, Dashti A. Chlorpyrifos Toxicity in Mouse Cultured Cerebellar Granule Neurons at Different Stages of Development: Additive Effect on Glutamate-Induced Excitotoxicity. CELL JOURNAL 2016; 18:464-72. [PMID: 27602329 PMCID: PMC5011335 DOI: 10.22074/cellj.2016.4575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 01/06/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Chlorpyrifos (CPF) is a neurotoxic organophosphorus (OP) insecticide. Its mechanism of action includes oxidative stress, excitotoxicity, and inhibition of the acetylcholinesterase enzyme (AChE). The aim of the present study is to investigate CPF toxicity in mature and immature cerebellar granule neurons (CGNs), as well as its effect on glutamate induced excitotoxicity. MATERIALS AND METHODS This study was an in vitro experimental study performed on mice cultured CGNs. Immature and mature neurons were exposed to different concentrations of CPF (1-1000 µM) and glutamate (10-600 µM) for 48 hours after which we used the MTT assay to measure cytotoxicity. Immature neurons had exposure to CPF for 5 days in order to evaluate the cytotoxic effect on developing neurons. Mature neurons received sub-lethal concentrations of CPF (10, 100 µM) combined with different concentrations of glutamate. AChE activity and reactive oxygen species (ROS) generation were assessed after treatments. RESULTS Immature CGNs had increased sensitivity to CPF toxicity compared to mature neurons. We observed significantly greater ROS production in immature compared to mature neurons, however AChE activity was more inhibited in mature neurons. Although CPF toxicity was not well correlated with AChE inhibition, it correlated well with ROS production. Glutamate toxicity was potentiated by sub-lethal concentration of CPF, however glutamate induced ROS production was not affected. The results suggested that CPF potentiated glutamate toxicity by mechanisms other than oxidative stress. CONCLUSION CPF toxicity differed in mature and immature neurons. Potentiated glutamate toxicity by CPF implied that CPF exposure might be a risk factor for neurodegenerative disease.
Collapse
Affiliation(s)
- Nahid Amani
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maliheh Soodi
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahram Daraei
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abolfazl Dashti
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
15
|
Linn CL. Retinal ganglion cell neuroprotection induced by activation of alpha7 nicotinic acetylcholine receptors. Neural Regen Res 2016; 11:918-9. [PMID: 27482217 PMCID: PMC4962586 DOI: 10.4103/1673-5374.184488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Cindy L Linn
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, USA
| |
Collapse
|
16
|
Wen D, Song W, Liu S, Tan X, Liu F. Upregulated expression of N-methyl-D-aspartate receptor 1 and nitric oxide synthase during form-deprivation myopia in guinea pigs. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:3819-3826. [PMID: 26097564 PMCID: PMC4466951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/21/2015] [Indexed: 06/04/2023]
Abstract
This study aimed to investigate the expression of N-methyl-D-aspartate receptor 1 (NMDAR1) and neuronal constitutive nitric oxide synthase (ncNOS) during form-deprivation myopia (FDM). FDM models were established in guinea pigs with facemasks. NMDAR1 expression in the retina was detected by immunohistochemistry and Western blot analysis. ncNOS mRNA expression was examined by in situ hybridization. cGMP content was measured by radioimmunoassay. In control group, NMDAR1 and ncNOS were expressed in binocular retinas, and there was no significant difference in NMDAR1 and ncNOS expression and cGMP content between the two eyes. However, NMDAR1 and ncNOS expression and cGMP content in the retina of FDM eyes were significantly higher than that of contralateral untreated eyes. Furthermore, ncNOS mRNA level and cGMP content was highly correlated. In conclusion, FDM upregulates the expression of NMDAR1 and ncNOS and increases cGMP content in the retina. NMDAR1/NO-cGMP pathway may contribute to abnormal visual signals during myopic progression.
Collapse
Affiliation(s)
- Dan Wen
- Department of Ophthalmology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Weitao Song
- Department of Ophthalmology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Shuangzhen Liu
- Department of Ophthalmology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Xingping Tan
- Department of Ophthalmology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Fei Liu
- Department of Infectious Disease, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| |
Collapse
|
17
|
Guerra-Álvarez M, Moreno-Ortega AJ, Navarro E, Fernández-Morales JC, Egea J, López MG, Cano-Abad MF. Positive allosteric modulation of alpha-7 nicotinic receptors promotes cell death by inducing Ca(2+) release from the endoplasmic reticulum. J Neurochem 2015; 133:309-19. [PMID: 25650007 DOI: 10.1111/jnc.13049] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 01/15/2015] [Accepted: 01/16/2015] [Indexed: 12/11/2022]
Abstract
Positive allosteric modulation of α7 isoform of nicotinic acetylcholine receptors (α7-nAChRs) is emerging as a promising therapeutic approach for central nervous system disorders such as schizophrenia or Alzheimer's disease. However, its effect on Ca(2+) signaling and cell viability remains controversial. This study focuses on how the type II positive allosteric modulator (PAM II) PNU120596 affects intracellular Ca(2+) signaling and cell viability. We used human SH-SY5Y neuroblastoma cells overexpressing α7-nAChRs (α7-SH) and their control (C-SH). We monitored cytoplasmic and endoplasmic reticulum (ER) Ca(2+) with Fura-2 and the genetically encoded cameleon targeting the ER, respectively. Nicotinic inward currents were measured using patch-clamp techniques. Viability was assessed using methylthiazolyl blue tetrazolium bromide or propidium iodide staining. We observed that in the presence of a nicotinic agonist, PNU120596 (i) reduced viability of α7-SH but not of C-SH cells; (ii) significantly increased inward nicotinic currents and cytosolic Ca(2+) concentration; (iii) released Ca(2+) from the ER by a Ca(2+) -induced Ca(2+) release mechanism only in α7-SH cells; (iv) was cytotoxic in rat organotypic hippocampal slice cultures; and, lastly, all these effects were prevented by selective blockade of α7-nAChRs, ryanodine receptors, or IP3 receptors. In conclusion, positive allosteric modulation of α7-nAChRs with the PAM II PNU120596 can lead to dysregulation of ER Ca(2+) , overloading of intracellular Ca(2+) , and neuronal cell death. This study focuses on how the type II positive allosteric modulator PNU120596 (PAM II PNU12) affects intracellular Ca(2+) signaling and cell viability. Using SH-SY5Y neuroblastoma cells overexpressing α7-nAChRs (α7-SH) and their control (C-SH), we find that PAM of α7-nAChRs with PNU120596: (i) increases inward calcium current (ICa ) and cytosolic Ca(2+) concentration ([Ca(2+) ]cyt ); (ii) releases Ca(2+) from the ER ([Ca(2+) ]ER ) by a Ca(2+) -induced Ca(2+) release mechanism; and (iv) reduces cell viability. These findings were corroborated in rat hippocampal organotypic cultures. [Ca(2+) ]cyt , cytosolic Ca(2+) concentration; [Ca(2+) ]ER , endoplasmic reticulum Ca(2+) concentration; α7 nAChR, α7 isoform of nicotinic acetylcholine receptors; α7-SH, SH-SY5Y stably overexpressing α7 nAChRs cells; C-SH, control SH-SY5Y cells; Nic, nicotine; PNU12, PNU120596.
Collapse
Affiliation(s)
- María Guerra-Álvarez
- Servicio de Farmacología Clínica, Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
18
|
Yamagishi R, Aihara M. Neuroprotective effect of astaxanthin against rat retinal ganglion cell death under various stresses that induce apoptosis and necrosis. Mol Vis 2014; 20:1796-805. [PMID: 25593507 PMCID: PMC4287717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 12/30/2014] [Indexed: 10/30/2022] Open
Abstract
PURPOSE Astaxanthin is a type of carotenoid known to have strong antioxidant effects. The purpose of this study was to investigate whether astaxanthin confers a neuroprotective effect against glutamate stress, oxidative stress, and hypoxia-induced apoptotic or necrotic cell death in primary cultures of rat retinal ganglion cells (RGCs). METHODS Purified rat RGCs were exposed to three kinds of stressors induced by 25 μM glutamate for 72 h, B27 medium without an antioxidant for 4 h, and a reduced oxygen level of 5% for 12 h. Each assay was repeated 12 times, with or without 1 nM, 10 nM, and 100 nM astaxanthin. The number of live RGCs was then counted using a cell viability assay. RGC viability in each condition was evaluated and compared with controls. In addition, we measured apoptosis and DNA damage. RESULTS We found that under glutamate stress, RGC viability was reduced to 58%. Cultures with 1 nM, 10 nM, and 100 nM astaxanthin showed an increase in RGC viability of 63%, 74%, and 84%, respectively. Under oxidative stress, RGC viability was reduced to 40%, and astaxanthin administration resulted in increased viability of 43%, 50%, and 67%, respectively. Under hypoxia, RGC viability was reduced to 66%, and astaxanthin administration resulted in a significant increase in viability to 67%, 77%, and 93%, respectively. These results indicate that 100 nM astaxanthin leads to a statistically significant increase in RGC viability under the three kinds of stressors tested, compared to controls (Dunnett's test, p<0.05). The apoptotic activity of RGCs under glutamate stress increased to 32%, but was reduced to 15% with 100 nM astaxanthin administration. Glutamate stress led to a 58% increase in DNA damage, which was reduced to 43% when cultured with 100 nM astaxanthin. Thus, 100 nM astaxanthin showed a statistically significant reduction in apoptosis and DNA damage in RGCs (Wilcoxon rank-sum test, p<0.05). CONCLUSIONS Our results suggest that astaxanthin has a neuroprotective effect against RGC death induced by glutamate stress, oxidative stress, and hypoxia, which induce apoptotic and necrotic cell death.
Collapse
Affiliation(s)
- Reiko Yamagishi
- Department of Ophthalmology, University of Tokyo School of Medicine, Tokyo, Japan
| | - Makoto Aihara
- Department of Ophthalmology, University of Tokyo School of Medicine, Tokyo, Japan,Shirato Eye Clinic, Tokyo, Japan
| |
Collapse
|
19
|
Iwamoto K, Birkholz P, Schipper A, Mata D, Linn DM, Linn CL. A nicotinic acetylcholine receptor agonist prevents loss of retinal ganglion cells in a glaucoma model. Invest Ophthalmol Vis Sci 2014; 55:1078-87. [PMID: 24458148 DOI: 10.1167/iovs.13-12688] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The purpose of this study was to analyze the neuroprotective effect of an α7 nAChR agonist, PNU-282987, using an in vivo model of glaucoma in Long Evans rats. METHODS One eye in each animal was surgically manipulated to induce glaucoma in control untreated animals and in animals that were treated with intravitreal injections of PNU-282987. To induce glaucoma-like conditions, 0.05 mL of 2 M NaCl was injected into the episcleral veins of right eyes in each rat to create scar tissue and increase intraocular pressure. The left eye in each rat acted as an internal control. One month following NaCl injection, rats were euthanized, retinas were removed, flatmounted, fixed, and nuclei were stained with cresyl violet or RGCs were immunostained with an antibody against Thy 1.1 or against Brn3a. Stained nuclei in the RGC layer and labeled RGCs in NaCl-injected retinas were counted and compared with cell counts from untreated retinas in the same animal. RESULTS NaCl injections into the episcleral veins caused a significant loss of cells by an average of 27.35% (± 2.12 SEM) in the RGC layer within 1 month after NaCl injection, which corresponded to a significant loss of RGCs. This loss of RGCs was eliminated if 5 μL of 100 μM PNU-282987 was injected into the right eye an hour before NaCl injection. CONCLUSIONS The results from this study support the hypothesis that the α7 agonist, PNU-282987, has a neuroprotective effect in the rat retina. PNU-282987 may be a viable candidate for future therapeutic treatments of glaucoma.
Collapse
Affiliation(s)
- Kazuhiro Iwamoto
- Western Michigan University, Department of Biological Sciences, Kalamazoo, Michigan
| | | | | | | | | | | |
Collapse
|
20
|
Sharifipour M, Izadpanah E, Nikkhoo B, Zare S, Abdolmaleki A, Hassanzadeh K, Moradi F, Hassanzadeh K. A new pharmacological role for donepezil: attenuation of morphine-induced tolerance and apoptosis in rat central nervous system. J Biomed Sci 2014; 21:6. [PMID: 24455992 PMCID: PMC3906771 DOI: 10.1186/1423-0127-21-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 01/20/2014] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Tolerance to the analgesic effect of opioids is a pharmacological phenomenon that occurs after their prolonged administration. It has been shown that morphine-induced tolerance is associated with apoptosis in the central nervous system and neuroprotective agents which prevented apoptosis signaling could attenuate tolerance to the analgesic effects. On the other hand donepezil, an acetylcholinesterase inhibitor, has been reported to have neuroprotective effects. Therefore in this study, the effect of systemic administration of donepezil on morphine-induced tolerance and apoptosis in the rat cerebral cortex and lumbar spinal cord was evaluated. Various groups of rats received morphine (ip) and different doses of donepezil (0, 0.5, 1, 1.5 mg/kg/day). Nociception was assessed using tail flick apparatus. Tail flick latency was recorded when the rat shook its tail. For apoptosis assay other groups of rats received the above treatment and apoptosis was evaluated by in situ terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL) method. RESULTS The results showed that administration of donepezil (0.5, 1, 1.5 mg/kg, ip) delayed the morphine tolerance for 9, 12 and 17 days, respectively. Furthermore pretreatment injection of donepezil attenuated the number of apoptotic cells in the cerebral cortex and lumbar spinal cord compared to the control group. CONCLUSION In conclusion, we found that systemic administration of donepezil attenuated morphine-induced tolerance and apoptosis in the rat cerebral cortex and lumbar spinal cord.
Collapse
Affiliation(s)
- Mozhdeh Sharifipour
- Department of Biology, Faculty of Basic Science, Urmia University, Urmia, Iran
| | - Esmaeal Izadpanah
- Cellular and Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Department of Physiology and Pharmacology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Bahram Nikkhoo
- Department of Pathology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Samad Zare
- Department of Biology, Faculty of Basic Science, Urmia University, Urmia, Iran
| | - Ali Abdolmaleki
- Science and Research Branch, Islamic Azad University, Hamedan, Iran
| | - Katayoun Hassanzadeh
- Department of Physiology and Pharmacology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Farshid Moradi
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Kambiz Hassanzadeh
- Cellular and Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Department of Physiology and Pharmacology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
21
|
Jang JY, Choi YW, Kim HN, Kim YR, Hong JW, Bae DW, Park SJ, Shin HK, Choi BT. Neuroprotective effects of a novel single compound 1-methoxyoctadecan-1-ol isolated from Uncaria sinensis in primary cortical neurons and a photothrombotic ischemia model. PLoS One 2014; 9:e85322. [PMID: 24416390 PMCID: PMC3885700 DOI: 10.1371/journal.pone.0085322] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 11/25/2013] [Indexed: 11/19/2022] Open
Abstract
We identified a novel neuroprotective compound, 1-methoxyoctadecan-1-ol, from Uncaria sinensis (Oliv.) Havil and investigated its effects and mechanisms in primary cortical neurons and in a photothrombotic ischemic model. In primary rat cortical neurons against glutamate-induced neurotoxicity, pretreatment with 1-methoxyoctadecan-1-ol resulted in significantly reduced neuronal death in a dose-dependent manner. In addition, treatment with 1-methoxyoctadecan-1-ol resulted in decreased neuronal apoptotic death, as assessed by nuclear morphological approaches. To clarify the neuroprotective mechanism of 1-methoxyoctadecan-1-ol, we explored the downstream signaling pathways of N-methyl-D-aspartate receptor (NMDAR) with calpain activation. Treatment with glutamate leads to early activation of NMDAR, which in turn leads to calpain-mediated cleavage of striatal-enriched protein tyrosine phosphatase (STEP) and subsequent activation of p38 mitogen activated protein kinase (MAPK). However, pretreatment with 1-methoxyoctadecan-1-ol resulted in significantly attenuated activation of GluN2B-NMDAR and a decrease in calpain-mediated STEP cleavage, leading to subsequent attenuation of p38 MAPK activation. We confirmed the critical role of p38 MAPK in neuroprotective effects of 1-methoxyoctadecan-1-ol using specific inhibitor SB203580. In the photothrombotic ischemic injury in mice, treatment with 1-methoxyoctadecan-1-ol resulted in significantly reduced infarct volume, edema size, and improved neurological function. 1-methoxyoctadecan-1-ol effectively prevents cerebral ischemic damage through down-regulation of calpain-mediated STEP cleavage and activation of p38 MAPK. These results suggest that 1-methoxyoctadecan-1-ol showed neuroprotective effects through down-regulation of calpain-mediated STEP cleavage with activation of GluN2B-NMDAR, and subsequent alleviation of p38 MAPK activation. In addition, 1-methoxyoctadecan-1-ol might be a useful therapeutic agent for brain disorder such as ischemic stroke.
Collapse
Affiliation(s)
- Ji Yeon Jang
- Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Young Whan Choi
- Department of Horticultural Bioscience, College of Natural Resource and Life Science, Pusan National University, Miryang, Gyeongnam, Republic of Korea
| | - Ha Neui Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Yu Ri Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Jin Woo Hong
- Division of Clinical Medicine 1, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Dong Won Bae
- Central Instrument Facility, Biomaterial Analytical Lab., Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Se Jin Park
- Department of Horticultural Bioscience, College of Natural Resource and Life Science, Pusan National University, Miryang, Gyeongnam, Republic of Korea
| | - Hwa Kyoung Shin
- Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- * E-mail: (BTC); (HKS)
| | - Byung Tae Choi
- Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- * E-mail: (BTC); (HKS)
| |
Collapse
|
22
|
Ortuño-Sahagún D, González RM, Verdaguer E, Huerta VC, Torres-Mendoza BM, Lemus L, Rivera-Cervantes MC, Camins A, Zárate CB. Glutamate excitotoxicity activates the MAPK/ERK signaling pathway and induces the survival of rat hippocampal neurons in vivo. J Mol Neurosci 2013; 52:366-77. [PMID: 24190281 DOI: 10.1007/s12031-013-0157-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 10/18/2013] [Indexed: 11/25/2022]
Abstract
Current knowledge concerning the molecular mechanisms of the cellular response to excitotoxic insults in neurodegenerative diseases is insufficient. Although glutamate (Glu) has been widely studied as the main excitatory neurotransmitter and principal excitotoxic agent, the neuroprotective response enacted by neurons is not yet completely understood. Some of the molecular participants have been revealed, but the signaling pathways involved in this protective response are just beginning to be identified. Here, we demonstrate in vivo that, in response to the cell damage and death induced by Glu excitotoxicity, neurons orchestrate a survival response through the extracellular signal-regulated kinase (ERK) signaling pathway by increasing ERK expression in the rat hippocampal (CA1) region, allowing increased neuronal survival. In addition, this protective response is specifically reversed by U0126, an ERK inhibitor, which promotes cell death only when it is administered together with Glu. Our findings demonstrate that the ERK signaling pathway has a neuroprotective role in the response to Glu-induced excitotoxicity in hippocampal neurons. Therefore, the ERK signaling pathway may be activated as a cellular response to excitotoxic injury to prevent damage and neural loss, representing a novel therapeutic target in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Daniel Ortuño-Sahagún
- Laboratorio de Desarrollo y Regeneración Neural, Instituto de Neurobiología, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Camino Ing. R. Padilla Sánchez, 2100, Las Agujas, Zapopan, 44600, Jalisco, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Swartz MM, Linn DM, Linn CL. Tropisetron as a neuroprotective agent against glutamate-induced excitotoxicity and mechanisms of action. Neuropharmacology 2013; 73:111-21. [PMID: 23727438 DOI: 10.1016/j.neuropharm.2013.05.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 04/23/2013] [Accepted: 05/07/2013] [Indexed: 11/30/2022]
Abstract
The objective of this study was to determine the neuroprotective role of tropisetron on retinal ganglion cells (RGCs) as well as to explore the possible mechanisms associated with alpha7 nAChR-induced neuroprotection. Adult pig RGCs were isolated from all other retinal tissue using a two-step panning technique. Once isolated, RGCs were cultured for 3 days under control untreated conditions, in the presence of 500 μM glutamate to induce excitotoxicity, and when tropisetron was applied before glutamate to induce neuroprotection. 500 μM glutamate decreased RGC survival by an average of 62% compared to control conditions. However, RGCs pretreated with 100 nM tropisetron before glutamate increased cell survival to an average of 105% compared to controls. Inhibition studies using the alpha7 nAChR antagonist, MLA (10 nM), support the hypothesis that tropisetron is an effective neuroprotective agent against glutamate-induced excitotoxicity; mediated by α7 nAChR activation. ELISA studies were performed to determine if signaling cascades normally associated with excitotoxicity and neuroprotection were up- or down-regulated after tropisetron treatment. Tropisetron had no discernible effects on pAkt levels but significantly decreased p38 MAPK levels associated with excitotoxicity from an average of 15 ng/ml to 6 ng/ml. Another mechanism shown to be associated with neuroprotection involves internalization of NMDA receptors. Double-labeled immunocytochemistry and electrophysiology studies provided further evidence that tropisetron caused internalization of NMDA receptor subunits. The findings of this study suggest that tropisetron could be an effective therapeutic agent for the treatment of degenerative disorders of the central nervous system that involves excitotoxicity.
Collapse
Affiliation(s)
- Michael M Swartz
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI 49008, USA
| | | | | |
Collapse
|
24
|
Iwamoto K, Mata D, Linn DM, Linn CL. Neuroprotection of rat retinal ganglion cells mediated through alpha7 nicotinic acetylcholine receptors. Neuroscience 2013; 237:184-98. [PMID: 23402849 DOI: 10.1016/j.neuroscience.2013.02.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 01/23/2013] [Accepted: 02/01/2013] [Indexed: 12/18/2022]
Abstract
Glutamate-induced excitotoxicity is thought to play an important role in several neurodegenerative diseases in the central nervous system (CNS). In this study, neuroprotection against glutamate-induced excitotoxicity was analyzed using acetylcholine (ACh), nicotine and the α7 specific nicotinic acetylcholine receptor (α7 nAChR) agonist, N-[(3R)-1-azabicyclo[2.2.2]oct-3-yl]-4-chlorobenzamide hydrochloride (PNU-282987), in cultured adult rat retinal neurons. Adult Long Evans rat retinas were dissociated and retinal ganglion cells (RGCs) were isolated from all other retinal tissue using a two-step panning technique. Once isolated, RGCs were cultured under various pharmacological conditions to demonstrate excitotoxicity and neuroprotection against excitotoxicity. After 3 days, RGCs were immunostained with antibodies against the glycoprotein, Thy 1.1, counted and cell survival was assessed relative to control untreated conditions. 500 μM glutamate induced excitotoxicity in large and small RGCs in an adult rat dissociated culture. After 3 days in culture with glutamate, the cell survival of large RGCs decreased by an average of 48.16% while the cell survival of small RGCs decreased by an average of 42.03%. Using specific glutamate receptor agonists and antagonists, we provide evidence that the excitotoxic response was mediated through α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainic acid (KA) and N-methyl-d-aspartate (NMDA) glutamate receptors through an apoptotic mechanism. However, the excitotoxic effect of glutamate on all RGCs was eliminated if cells were cultured for an hour with 10 μM ACh, 100 μM nicotine or 100 nM of the α7 nAChR agonist, PNU-282987, before the glutamate insult. Inhibition studies using 10nM methyllycaconitine (MLA) or α-bungarotoxin (α-Bgt) supported the hypothesis that neuroprotection against glutamate-induced excitotoxicity on rat RGCs was mediated through α7 nAChRs. In immunocytochemical studies, double-labeled experiments using antibodies against Thy 1.1 and α7 nAChR subunits demonstrated that both large and small RGCs contained α7 nAChR subunits. The data presented in this study support the hypothesis that ACh and nicotinic acetylcholine receptor (nAChR) agonists provide neuroprotection against glutamate-induced excitotoxicity in adult rat RGCs through activation of α7 nAChR subunits. These studies lay the groundwork required for analyzing the effect of specific α7 nAChR agonists using in vivo models of excitotoxicity. Understanding the type of ACh receptors involved in neuroprotection in the rat retina could ultimately lead to therapeutic treatment for any CNS disease that involves excitotoxicity.
Collapse
Affiliation(s)
- K Iwamoto
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI 49008, USA
| | | | | | | |
Collapse
|
25
|
Abstract
Retinal ischemia is a very useful model to study the impact of various cell death pathways, such as apoptosis and necrosis, in the ischemic retina. However, it is important to note that the retina is formed as an outpouching of the diencephalon and is part of the central nervous system. As such, the cell death pathways initiated in response to ischemic damage in the retina reflect those found in other areas of the central nervous system undergoing similar trauma. The retina is also more accessible than other areas of the central nervous system, thus making it a simpler model to work with and study. By utilizing the retinal model, we can greatly increase our knowledge of the cell death processes initiated by ischemia which lead to degeneration in the central nervous system. This paper examines work that has been done so far to characterize various aspects of cell death in the retinal ischemia model, such as various pathways which are activated, and the role neurotrophic factors, and discusses how these are relevant to the treatment of ischemic damage in both the retina and the greater central nervous system.
Collapse
|
26
|
Mehta A, Prabhakar M, Kumar P, Deshmukh R, Sharma PL. Excitotoxicity: bridge to various triggers in neurodegenerative disorders. Eur J Pharmacol 2012; 698:6-18. [PMID: 23123057 DOI: 10.1016/j.ejphar.2012.10.032] [Citation(s) in RCA: 474] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 09/26/2012] [Accepted: 10/06/2012] [Indexed: 12/13/2022]
Abstract
Glutamate is one of the most prominent neurotransmitter in the body, present in over 50% of nervous tissue and plays an important role in neuronal excitation. This neuronal excitation is short-lived and is followed by depression. Multiple abnormal triggers such as energy deficiency, oxidative stress, mitochondrial dysfunction, calcium overload, etc can lead to aberration in neuronal excitation process. Such an aberration, serves as a common pool or bridge between abnormal triggers and deleterious signaling processes with which central neurons cannot cope up, leading to death. Excitotoxicity is the pathological process by which nerve cells are damaged and killed by excessive stimulation by neurotransmitters such as glutamate and similar substances. Such excitotoxic neuronal death has been implicated in spinal cord injury, stroke, traumatic brain injury, hearing loss and in neurodegenerative diseases of the central nervous system such as stroke, epilepsy, multiple sclerosis, Alzheimer disease, Amyltropic lateral sclerosis, Parkinson's disease, Huntington disease and alcohol withdrawal. This review mainly emphasizes the triggering events which sustain neuronal excitation, role of calcium, mitochondrial dysfunction, ROS, NO, chloride homeostasis and eicosanoids pathways. Further, a brief introduction about the recent research occurring in the treatment of various neurodegenerative diseases, including a summary of the presumed physiologic mechanisms behind the pharmacology of these disorders.
Collapse
Affiliation(s)
- Ankita Mehta
- Neuropharmacology Division, ISF College of Pharmacy, Ferozpur Road, Ghal Kalan, Moga 142 001, Punjab, India
| | | | | | | | | |
Collapse
|
27
|
Wu XM, Qian ZM, Zhu L, Du F, Yung WH, Gong Q, Ke Y. Neuroprotective effect of ligustilide against ischaemia-reperfusion injury via up-regulation of erythropoietin and down-regulation of RTP801. Br J Pharmacol 2012; 164:332-43. [PMID: 21410687 DOI: 10.1111/j.1476-5381.2011.01337.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Ligustilide, the main lipophilic component of Danggui, has been reported to protect the brain against ischaemic injury. However, the mechanisms are unknown. Here, we investigated the roles of erythropoietin (EPO) and the stress-induced protein RTP801 in neuroprotection provided by ligustilide against ischaemia-reperfusion (I/R) damage to the brain. EXPERIMENTAL APPROACH The efficacy of ligustilide against I/R damage was assessed by neurological deficit, infarct volume and cell viability, using the middle cerebral artery occlusion model in rats in vivo and rat cultured neurons in vitro. EPO and RTP801 were analysed by Western blot. Over-expression of RTP801 was achieved by transfection of an expression plasmid. KEY RESULTS Ligustilide decreased the neurological deficit score, infarct volume and RTP801 expression and increased EPO transcription in I/R rats, and increased cell viability and EPO and decreased LDH release and RTP801 in I/R neurons. Also, ligustilide increased ERK phosphorylation (p-ERK). The positive effects of ligustilide on p-ERK, cell viability and EPO were blocked by PD98059, but not LY294002 and SB203580. In addition, transfection of SH-SY5Y cells with RTP801 plasmid increased RTP801 and LDH release, while ligustilide inhibited the effects of transfection on RTP801 expression and also increased cell viability. CONCLUSION AND IMPLICATIONS Ligustilide exerts neuroprotective effects against I/R injury by promoting EPO transcription via an ERK signalling pathway and inhibiting RTP801 expression, This compound could be developed into a therapeutic agent to prevent and treat ischaemic disorders.
Collapse
Affiliation(s)
- Xiao-mei Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | | | | | | | | | | | | |
Collapse
|
28
|
Nicotinic stimulation induces Tristetraprolin over-production and attenuates inflammation in muscle. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:368-78. [DOI: 10.1016/j.bbamcr.2011.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 10/27/2011] [Accepted: 11/02/2011] [Indexed: 01/11/2023]
|
29
|
Uteshev VV. α7 nicotinic ACh receptors as a ligand-gated source of Ca(2+) ions: the search for a Ca(2+) optimum. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:603-38. [PMID: 22453962 DOI: 10.1007/978-94-007-2888-2_27] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The spatiotemporal distribution of cytosolic Ca(2+) ions is a key determinant of neuronal behavior and survival. Distinct sources of Ca(2+) ions including ligand- and voltage-gated Ca(2+) channels contribute to intracellular Ca(2+) homeostasis. Many normal physiological and therapeutic neuronal functions are Ca(2+)-dependent, however an excess of cytosolic Ca(2+) or a lack of the appropriate balance between Ca(2+) entry and clearance may destroy cellular integrity and cause cellular death. Therefore, the existence of optimal spatiotemporal patterns of cytosolic Ca(2+) elevations and thus, optimal activation of ligand- and voltage-gated Ca(2+) ion channels are postulated to benefit neuronal function and survival. Alpha7 nicotinic -acetylcholine receptors (nAChRs) are highly permeable to Ca(2+) ions and play an important role in modulation of neurotransmitter release, gene expression and neuroprotection in a variety of neuronal and non-neuronal cells. In this review, the focus is placed on α7 nAChR-mediated currents and Ca(2+) influx and how this source of Ca(2+) entry compares to NMDA receptors in supporting cytosolic Ca(2+) homeostasis, neuronal function and survival.
Collapse
Affiliation(s)
- Victor V Uteshev
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX 76107, USA.
| |
Collapse
|
30
|
Ko ML, Chen CF, Peng PH, Peng YH. Simvastatin upregulates Bcl-2 expression and protects retinal neurons from early ischemia/reperfusion injury in the rat retina. Exp Eye Res 2011; 93:580-5. [DOI: 10.1016/j.exer.2011.07.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 04/23/2011] [Accepted: 07/06/2011] [Indexed: 01/13/2023]
|
31
|
Cheyuo C, Jacob A, Wu R, Zhou M, Coppa GF, Wang P. The parasympathetic nervous system in the quest for stroke therapeutics. J Cereb Blood Flow Metab 2011; 31:1187-95. [PMID: 21364605 PMCID: PMC3099641 DOI: 10.1038/jcbfm.2011.24] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Stroke is a devastating neurovascular disease with limited therapeutic options. The pathogenesis of stroke involves complex interrelated molecular mechanisms including excitotoxicity, oxidative and nitrosative stress, cortical spreading depolarizations, inflammation, necrosis, and apoptosis. Successful development of stroke therapeutics depends on understanding these molecular mechanisms and how to counteract them to limit tissue damage during stroke. Activation of the parasympathetic nervous system (PNS) has been shown to antagonize a multiplicity of pathologic mechanisms. Elements of parasympathetic activation such as vagus nerve stimulation have already been used successfully in treating brain disorders such as epilepsy and depression. This review discusses the anatomical basis and molecular mechanisms involved in activation of the PNS, and assesses the strength of available evidence for the further development of this modality into a stroke therapy.
Collapse
Affiliation(s)
- Cletus Cheyuo
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | | | | | | | |
Collapse
|
32
|
Shen H, Kihara T, Hongo H, Wu X, Kem WR, Shimohama S, Akaike A, Niidome T, Sugimoto H. Neuroprotection by donepezil against glutamate excitotoxicity involves stimulation of alpha7 nicotinic receptors and internalization of NMDA receptors. Br J Pharmacol 2010; 161:127-39. [PMID: 20718745 DOI: 10.1111/j.1476-5381.2010.00894.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Glutamate excitotoxicity may be involved in ischaemic injury to the CNS and some neurodegenerative diseases, such as Alzheimer's disease. Donepezil, an acetylcholinesterase (AChE) inhibitor, exerts neuroprotective effects. Here we demonstrated a novel mechanism underlying the neuroprotection induced by donepezil. EXPERIMENTAL APPROACH Cell damage in primary rat neuron cultures was quantified by lactate dehydrogenase release. Morphological changes associated with neuroprotective effects of nicotine and AChE inhibitors were assessed by immunostaining. Cell surface levels of the glutamate receptor sub-units, NR1 and NR2A, were analyzed using biotinylation. Immunoblot was used to measure protein levels of cleaved caspase-3, total NR1, total NR2A and phosphorylated NR1. Immunoprecipitation was used to measure association of NR1 with the post-synaptic protein, PSD-95. Intracellular Ca(2+) concentrations were measured with fura 2-acetoxymethylester. Caspase 3-like activity was measured using enzyme substrate, 7-amino-4-methylcoumarin (AMC)-DEVD. KEY RESULTS Levels of NR1, a core subunit of the NMDA receptor, on the cell surface were significantly reduced by donepezil. In addition, glutamate-mediated Ca(2+) entry was significantly attenuated by donepezil. Methyllycaconitine, an inhibitor of alpha7 nicotinic acetylcholine receptors (nAChR), inhibited the donepezil-induced attenuation of glutamate-mediated Ca(2+) entry. LY294002, a phosphatidyl inositol 3-kinase (PI3K) inhibitor, had no effect on attenuation of glutamate-mediated Ca(2+) entry induced by donepezil. CONCLUSIONS AND IMPLICATIONS Decreased glutamate toxicity through down-regulation of NMDA receptors, following stimulation of alpha7 nAChRs, could be another mechanism underlying neuroprotection by donepezil, in addition to up-regulating the PI3K-Akt cascade or defensive system.
Collapse
Affiliation(s)
- H Shen
- Department of Neuroscience for Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Sokolowska P, Passemard S, Mok A, Schwendimann L, Gozes I, Gressens P. Neuroprotective effects of NAP against excitotoxic brain damage in the newborn mice: implications for cerebral palsy. Neuroscience 2010; 173:156-68. [PMID: 21073926 DOI: 10.1016/j.neuroscience.2010.10.074] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 10/23/2010] [Accepted: 10/27/2010] [Indexed: 10/18/2022]
Abstract
Activity-dependent neuroprotective protein (ADNP) was shown to be essential for embryogenesis and brain development while NAP, an active motif of ADNP, is neuroprotective in a broad range of neurodegenerative disorders. In the present study, we examined the protective potential of ADNP/NAP in a mouse model of excitotoxic brain lesion mimicking brain damage associated with cerebral palsy. We demonstrated that NAP had a potent neuroprotective effect against ibotenate-induced excitotoxic damage in the cortical plate and the white matter of P5 mice, and moderate against brain lesions of P0 mice. In contrast, endogenous ADNP appears not to be involved in the response to excitotoxic challenge in the studied model. Our findings further show that NAP reduced the number of apoptotic neurons through activation of PI-3K/Akt pathway in the cortical plate or both PI-3K/Akt and MAPK/MEK1 kinases in the white matter. In addition, NAP prevented ibotenate-induced loss of pre-oligodendrocytes without affecting the number of astrocytes or activated microglia around the site of injection. These findings indicate that protective actions of NAP are mediated by triggering transduction pathways that are crucial for neuronal and oligodendroglial survival, thus, NAP might be a promising therapeutic agent for treating developing brain damage.
Collapse
|
34
|
Brandt SK, Weatherly ME, Ware L, Linn DM, Linn CL. Calcium preconditioning triggers neuroprotection in retinal ganglion cells. Neuroscience 2010; 172:387-97. [PMID: 21044663 DOI: 10.1016/j.neuroscience.2010.10.071] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 10/21/2010] [Accepted: 10/26/2010] [Indexed: 10/18/2022]
Abstract
In the mammalian retina, excitotoxicity has been shown to be involved in apoptotic retinal ganglion cell (RGC) death and is associated with certain retinal disease states including glaucoma, diabetic retinopathy and retinal ischemia. Previous studies from this lab [Wehrwein E, Thompson SA, Coulibaly SF, Linn DM, Linn CL (2004) Invest Ophthalmol Vis Sci 45:1531-1543] have demonstrated that acetylcholine (ACh) and nicotine protects against glutamate-induced excitotoxicity in isolated adult pig RGCs through nicotinic acetylcholine receptors (nAChRs). Activation of nAChRs in these RGCs triggers cell survival signaling pathways and inhibits apoptotic enzymes [Asomugha CO, Linn DM, Linn CL (2010) J Neurochem 112:214-226]. However, the link between binding of nAChRs and activation of neuroprotective pathways is unknown. In this study, we examine the hypothesis that calcium permeation through nAChR channels is required for ACh-induced neuroprotection against glutamate-induced excitotoxicity in isolated pig RGCs. RGCs were isolated from other retinal tissue using a two step panning technique and cultured for 3 days under different conditions. In some studies, calcium imaging experiments were performed using the fluorescent calcium indicator, fluo-4, and demonstrated that calcium permeates the nAChR channels located on pig RGCs. In other studies, the extracellular calcium concentration was altered to determine the effect on nicotine-induced neuroprotection. Results support the hypothesis that calcium is required for nicotine-induced neuroprotection in isolated pig RGCs. Lastly, studies were performed to analyze the effects of preconditioning on glutamate-induced excitotoxicity and neuroprotection. In these studies, a preconditioning dose of calcium was introduced to cells using a variety of mechanisms before a large glutamate insult was applied to cells. Results from these studies support the hypothesis that preconditioning cells with a relatively low level of calcium before an excitotoxic insult leads to neuroprotection. In the future, these results could provide important information concerning therapeutic agents developed to combat various diseases involved with glutamate-induced excitotoxicity.
Collapse
Affiliation(s)
- S K Brandt
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| | | | | | | | | |
Collapse
|