1
|
Tauskela JS, Brunette E, Aylsworth A, Zhao X. Neuroprotection against supra-lethal 'stroke in a dish' insults by an anti-excitotoxic receptor antagonist cocktail. Neurochem Int 2022; 158:105381. [PMID: 35764225 DOI: 10.1016/j.neuint.2022.105381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/07/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
The goal of this study was to identify cocktails of drugs able to protect cultured rodent cortical neurons against increasing durations of oxygen-glucose deprivation (OGD). As expected, a cocktail composed of an NMDA and AMPA receptor antagonists and a voltage gated Ca2+ channel blocker (MK-801, CNQX and nifedipine, respectively) provided complete neuroprotection against mild OGD. Increasingly longer durations of OGD necessitated increasing the doses of MK-801 and CNQX, until these cocktails ultimately failed to provide neuroprotection against supra-lethal OGD, even at maximal drug concentrations. Surprisingly, supplementation of any of these cocktails with blockers of TRPM7 channels for increasing OGD durations was not neuroprotective, unless these blockers possessed the ability to inhibit NMDA receptors. Supplementation of the maximally effective cocktail with other NMDA receptor antagonists augmented neuroprotection, suggesting insufficient NMDAR blockade by MK-801. Substitution of MK-801 in cocktails with high concentrations of a glycine site NMDA receptor antagonist caused the greatest improvements in neuroprotection, with the more potent SM-31900 superior to L689,560. Substitution of CQNX in cocktails with AMPA receptor antagonists at high concentrations also improved neuroprotection, particularly with the combination of SYM 2206 and NBQX. The most neuroprotective cocktail was thus composed of SM-31900, SYM2206, NBQX, nifedipine and the antioxidant trolox. Thus, the cumulative properties of antagonist potency and concentration in a cocktail dictate neuroprotective efficacy. The central target of supra-lethal OGD is excitotoxicity, which must be blocked to the greatest extent possible to minimize ion influx.
Collapse
Affiliation(s)
- Joseph S Tauskela
- National Research Council of Canada, Human Health Therapeutics, Building M-54, 1200 Montreal Road, Ottawa, ON, Canada, K1A 0R6.
| | - Eric Brunette
- National Research Council of Canada, Human Health Therapeutics, Building M-54, 1200 Montreal Road, Ottawa, ON, Canada, K1A 0R6
| | - Amy Aylsworth
- National Research Council of Canada, Human Health Therapeutics, Building M-54, 1200 Montreal Road, Ottawa, ON, Canada, K1A 0R6
| | - Xigeng Zhao
- National Research Council of Canada, Human Health Therapeutics, Building M-54, 1200 Montreal Road, Ottawa, ON, Canada, K1A 0R6
| |
Collapse
|
2
|
Intracellular Signaling. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
3
|
Electrophysiological- and Neuropharmacological-Based Benchmarking of Human Induced Pluripotent Stem Cell-Derived and Primary Rodent Neurons. Stem Cell Rev Rep 2021; 18:259-277. [PMID: 34687385 DOI: 10.1007/s12015-021-10263-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2021] [Indexed: 12/15/2022]
Abstract
Human induced pluripotent stem cell (iPSC)-derived neurons are of interest for studying neurological disease mechanisms, developing potential therapies and deepening our understanding of the human nervous system. However, compared to an extensive history of practice with primary rodent neuron cultures, human iPSC-neurons still require more robust characterization of expression of neuronal receptors and ion channels and functional and predictive pharmacological responses. In this study, we differentiated human amniotic fluid-derived iPSCs into a mixed population of neurons (AF-iNs). Functional assessments were performed by evaluating electrophysiological (patch-clamp) properties and the effect of a panel of neuropharmacological agents on spontaneous activity (multi-electrode arrays; MEAs). These electrophysiological data were benchmarked relative to commercially sourced human iPSC-derived neurons (CNS.4U from Ncardia), primary human neurons (ScienCell™) and primary rodent cortical/hippocampal neurons. Patch-clamp whole-cell recordings showed that mature AF-iNs generated repetitive firing of action potentials in response to depolarizations, similar to that of primary rodent cortical/hippocampal neurons, with nearly half of the neurons displaying spontaneous post-synaptic currents. Immunochemical and MEA-based analyses indicated that AF-iNs were composed of functional glutamatergic excitatory and inhibitory GABAergic neurons. Principal component analysis of MEA data indicated that human AF-iN and rat neurons exhibited distinct pharmacological and electrophysiological properties. Collectively, this study establishes a necessary prerequisite for AF-iNs as a human neuron culture model suitable for pharmacological studies.
Collapse
|
4
|
Resilience of network activity in preconditioned neurons exposed to 'stroke-in-a-dish' insults. Neurochem Int 2021; 146:105035. [PMID: 33798645 DOI: 10.1016/j.neuint.2021.105035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 11/22/2022]
Abstract
Exposing cultured cortical neurons to stimulatory agents - the K+ channel blocker 4-aminopyridine (4-ap), and the GABAA receptor antagonist bicuculline (bic) - for 48 h induces down-regulated synaptic scaling, and preconditions neurons to withstand subsequent otherwise lethal 'stroke-in-a-dish' insults; however, the degree to which usual neuronal function remains is unknown. As a result, multi-electrode array and patch-clamp electrophysiological techniques were employed to characterize hallmarks of spontaneous synaptic activity over a 12-day preconditioning/insult experiment. Spiking frequency increased 8-fold immediately upon 4-ap/bic treatment but declined within the 48 h treatment window to sub-baseline levels that persisted long after washout. Preconditioning resulted in key markers of network activity - spiking frequency, bursting and avalanches - being impervious to an insult. Surprisingly, preconditioning resulted in higher peak NMDA mEPSC amplitudes, resulting in a decrease in the ratio of AMPA:NMDA mEPSC currents, suggesting a relative increase in synaptic NMDA receptors. An investigation of a broad mRNA panel of excitatory and inhibitory signaling mediators indicated preconditioning rapidly up-regulated GABA synthesis (GAD67) and BDNF, followed by up-regulation of neuronal activity-regulated pentraxin and down-regulation of presynaptic glutamate release (VGLUT1). Preconditioning also enhanced surface expression of GLT-1, which persisted following an insult. Overall, preconditioning resulted in a reduced spiking frequency which was impervious to subsequent exposure to 'stroke-in-a-dish' insults, a phenotype initiated predominantly by up-regulation of inhibitory neurotransmission, a lower neuronal postsynaptic AMPA: NMDA receptor ratio, and trafficking of GLT-1 to astrocyte plasma membranes.
Collapse
|
5
|
Singh A, Chow O, Jenkins S, Zhu L, Rose E, Astbury K, Chen R. Characterizing Ischaemic Tolerance in Rat Pheochromocytoma (PC12) Cells and Primary Rat Neurons. Neuroscience 2020; 453:17-31. [PMID: 33246056 DOI: 10.1016/j.neuroscience.2020.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/16/2022]
Abstract
Preconditioning tissue with sublethal ischaemia or hypoxia can confer tolerance (protection) against subsequent ischaemic challenge. In vitro ischaemic preconditioning (IPC) is typically achieved through oxygen glucose deprivation (OGD), whereas hypoxic preconditioning (HPC) involves oxygen deprivation (OD) alone. Here, we report the effects of preconditioning of OGD, OD or glucose deprivation (GD) in ischaemic tolerance models with PC12 cells and primary rat neurons. PC12 cells preconditioned (4 h) with GD or OGD, but not OD, prior to reperfusion (24 h) then ischaemic challenge (OGD 6 h), showed greater mitochondrial activity, reduced cytotoxicity and decreased apoptosis, compared to sham preconditioned PC12 cells. Furthermore, 4 h preconditioning with reduced glucose (0.565 g/L, reduced from 4.5 g/L) conferred protective effects, but not for higher concentrations (1.125 or 2.25 g/L). Preconditioning (4 h) with OGD, but not OD or GD, induced stabilization of hypoxia inducible factor 1α (HIF1α) and upregulation of HIF1 downstream genes (Vegf, Glut1, Pfkfb3 and Ldha). In primary rat neurons, only OGD preconditioning (4 h) conferred neuroprotection. OGD preconditioning (4 h) induced stabilization of HIF1α and upregulation of HIF1 downstream genes (Vegf, Phd2 and Bnip3). In conclusion, OGD preconditioning (4 h) followed by 24 h reperfusion induced ischaemic tolerance (against OGD, 6 h) in both PC12 cells and primary rat neurons. The OGD preconditioning protection is associated with HIF1α stabilization and upregulation of HIF1 downstream gene expression. GD preconditioning (4 h) leads to protection in PC12 cells, but not in neurons. This GD preconditioning-induced protection was not associated with HIF1α stabilization.
Collapse
Affiliation(s)
- Ayesha Singh
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK.
| | - Oliver Chow
- Department of Molecular, Cellular, Developmental Biology, University of Colorado, Boulder, CO 80302, USA
| | - Stuart Jenkins
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK.
| | - Lingling Zhu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Emily Rose
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK.
| | - Katherine Astbury
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK.
| |
Collapse
|
6
|
Liang W, Lin C, Yuan L, Chen L, Guo P, Li P, Wang W, Zhang X. Preactivation of Notch1 in remote ischemic preconditioning reduces cerebral ischemia-reperfusion injury through crosstalk with the NF-κB pathway. J Neuroinflammation 2019; 16:181. [PMID: 31526384 PMCID: PMC6747758 DOI: 10.1186/s12974-019-1570-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 08/29/2019] [Indexed: 12/21/2022] Open
Abstract
Background Remote ischemic preconditioning (RIPC) initiates endogenous protective pathways in the brain from a distance and represents a new, promising paradigm in neuroprotection against cerebral ischemia-reperfusion (I/R) injury. However, the underlying mechanism of RIPC-mediated cerebral ischemia tolerance is complicated and not well understood. We reported previously that preactivation of Notch1 mediated the neuroprotective effects of cerebral ischemic preconditioning in rats subjected to cerebral I/R injury. The present study seeks to further explore the role of crosstalk between the Notch1 and NF-κB signaling pathways in the process of RIPC-induced neuroprotection. Methods Middle cerebral artery occlusion and reperfusion (MCAO/R) in adult male rats and oxygen-glucose deprivation and reoxygenation (OGD/R) in primary hippocampal neurons were used as models of I/R injury in vivo and in vitro, respectively. RIPC was induced by a 3-day procedure with 4 cycles of 5 min of left hind limb ischemia followed by 5 min of reperfusion each day before MCAO/R. Intracerebroventricular DAPT injection and sh-Notch1 lentivirus interference were used to inhibit the Notch1 signaling pathway in vivo and in vitro, respectively. After 24 h of reperfusion, neurological deficit scores, infarct volume, neuronal apoptosis, and cell viability were assessed. The protein expression levels of NICD, Hes1, Phospho-IKKα/β (p-IKK α/β), Phospho-NF-κB p65 (p-NF-κB p65), Bcl-2, and Bax were assessed by Western blotting. Results RIPC significantly improved neurological scores and reduced infarct volume and neuronal apoptosis in rats subjected to I/R injury. OGD preconditioning significantly reduced neuronal apoptosis and improved cell viability after I/R injury on days 3 and 7 after OGD/R. However, the neuroprotective effect was reversed by DAPT in vivo and attenuated by Notch1-RNAi in vitro. RIPC significantly upregulated the expression of proteins related to the Notch1 and NF-κB pathways. NF-κB signaling pathway activity was suppressed by a Notch1 signaling pathway inhibitor and Notch1-RNAi. Conclusions The neuroprotective effect of RIPC against cerebral I/R injury was associated with preactivation of the Notch1 and NF-κB pathways in neurons. The NF-κB pathway is a downstream target of the Notch1 pathway in RIPC and helps protect focal cerebral I/R injury.
Collapse
Affiliation(s)
- Weidong Liang
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Chunshui Lin
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Liuqing Yuan
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Li Chen
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Peipei Guo
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ping Li
- Department of Anesthesia, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Wei Wang
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xin Zhang
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
7
|
Xie X, Peng L, Zhu J, Zhou Y, Li L, Chen Y, Yu S, Zhao Y. miR-145-5p/Nurr1/TNF-α Signaling-Induced Microglia Activation Regulates Neuron Injury of Acute Cerebral Ischemic/Reperfusion in Rats. Front Mol Neurosci 2017; 10:383. [PMID: 29209166 PMCID: PMC5702297 DOI: 10.3389/fnmol.2017.00383] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/03/2017] [Indexed: 01/03/2023] Open
Abstract
Nurr1 is a member of the nuclear receptor 4 family of orphan nuclear receptors that is decreased in inflammatory responses and leads to neurons death in Parkinson’s disease. Abnormal expression of Nurr1 have been attributed to various signaling pathways, but little is known about microRNAs (miRNAs) regulation of Nurr1 in ischemia/reperfusion injury. To investigate the post transcriptional regulatory networks of Nurr1, we used a miRNA screening approach and identified miR-145-5p as a putative regulator of Nurr1. By using computer predictions, we identified and confirmed a miRNA recognition element in the 3′UTR of Nurr1 that was responsible for miR-145-5p-mediated suppression. We next demonstrated that overexpression of Nurr1 inhibited TNF-α expression in microglia by trans-repression and finally attenuated ischemia/reperfusion-induced inflammatory and cytotoxic response of neurons. Results of further in vivo study revealed that anti-miR-145-5p administration brought about increasing expression of Nurr1 and reduction of infarct volume in acute cerebral ischemia. Administration of anti-miR-145-5p promotes neurological outcome of rats post MCAO/R. It might be an effective therapeutic strategy to relieve neurons injury upon ischemia/reperfusion of rats through interrupting the axis signaling of miR-145-5p- Nurr1-TNF-α in acute phase.
Collapse
Affiliation(s)
- Xuemei Xie
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Li Peng
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Jin Zhu
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Yang Zhou
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Lingyu Li
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Yanlin Chen
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Shanshan Yu
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Zhu K, He Q, Li L, Zhao Y, Zhao J. Silencing thioredoxin1 exacerbates damage of astrocytes exposed to OGD/R by aggravating apoptosis through the Actin-Ras2-cAMP-PKA pathway. Int J Neurosci 2017; 128:512-519. [PMID: 29073813 DOI: 10.1080/00207454.2017.1398159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE OF THE STUDY Induction of endogenous antioxidants is one of the key molecular mechanisms of cell resistance to hypoxia/ischemia. Thioredoxin1 (Trx1) is a small multifunctional ubiquitous antioxidant with redox-active dithiol and plays an important role in cell apoptosis through mitochondrial apoptosis pathways. The specific role of Trx1 in ischemia-reperfusion induced astrocyte apoptosis, however, remains unclear. MATERIALS AND METHODS In this study, we investigated the effect of Trx1 on apoptosis of astrocyte using an in vitro oxygen-glucose deprivation/reoxygenation (OGD/R) model which mimics ischemic/reperfusion conditions in vivo. The astrocytes prepared from newborn Sprague-Dawley rats were exposed to OGD for 4 h followed by reoxygenation for 24 h. Next, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was used to assess cell viability while cell damage was assessed by lactate dehydrogenase (LDH). RESULTS We found that OGD/R increased cell death as well as the expression of Trx1 and that the interference of Trx1 further aggravated astrocyte damage under OGD/R condition. Furthermore, we detected an increase in the intracellular expressions of Ras2, cAMP, and PKA under OGD/R condition, which paralleled cell injury. CONCLUSIONS Notably, the deletion of Trx1 exacerbated astrocyte apoptosis via the Ras2-cAMP-PKA signaling pathway. We concluded that Trx1 protects astrocytes against apoptotic injury induced by OGD/R, and this protective effect may be partly related to the Ras2-cAMP-PKA signaling pathway.
Collapse
Affiliation(s)
- Kunting Zhu
- a Department of Pathology , The First People's Hospital of Yibin , Yibin , Sichuan , People's Republic of China
| | - Qi He
- b Department of Pathophysiology , Chongqing Medical University , Chongqing , People's Republic of China.,c Institute of Neuroscience , Chongqing Medical University , Chongqing , PR China
| | - Lingyu Li
- c Institute of Neuroscience , Chongqing Medical University , Chongqing , PR China.,d Department of Pathology , Chongqing Medical University , Chongqing , People's Republic of China
| | - Yong Zhao
- c Institute of Neuroscience , Chongqing Medical University , Chongqing , PR China.,d Department of Pathology , Chongqing Medical University , Chongqing , People's Republic of China
| | - Jing Zhao
- b Department of Pathophysiology , Chongqing Medical University , Chongqing , People's Republic of China.,c Institute of Neuroscience , Chongqing Medical University , Chongqing , PR China
| |
Collapse
|
9
|
Liu R, Wang H, Xu B, Chen W, Turlova E, Dong N, Sun CLF, Lu Y, Fu H, Shi R, Barszczyk A, Yang D, Jin T, Mannucci E, Feng ZP, Sun HS. Cerebrovascular Safety of Sulfonylureas: The Role of KATP Channels in Neuroprotection and the Risk of Stroke in Patients With Type 2 Diabetes. Diabetes 2016; 65:2795-809. [PMID: 27207539 DOI: 10.2337/db15-1737] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/11/2016] [Indexed: 11/13/2022]
Abstract
Sulfonylureas are ATP-sensitive potassium (KATP) channel blockers commonly used in the treatment of type 2 diabetes mellitus (T2DM). Activation of KATP channels plays a neuroprotective role in ischemia; thus, whether sulfonylureas affect the outcomes of stroke in patients with T2DM needs to be further studied. In our study, streptozotocin (STZ)-induced diabetic mice subjected to transient middle cerebral artery occlusion (MCAO) showed larger areas of brain damage and poorer behavioral outcomes. Blocking the KATP channel by tolbutamide increased neuronal injury induced by oxygen-glucose deprivation (OGD) in vitro and permanent MCAO (pMCAO) in vivo. Activating the KATP channel by diazoxide reduced the effects of both the OGD and pMCAO. Western blot analysis in STZ mouse brains indicated an early increase in protein levels of N-methyl-d-aspartate receptor 2B and postsynaptic density protein-95, followed by a decrease in phosphorylation of glycogen synthase kinase 3β. Our systematic meta-analysis indicated that patients with T2DM treated with sulfonylureas had a higher odds ratio for stroke morbidity than those who received comparator drugs. Taken together, these results suggest that sulfonylurea treatment in patients with T2DM may inhibit the neuroprotective effects of KATP channels and increase the risk of stroke.
Collapse
Affiliation(s)
- Rui Liu
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Haitao Wang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Baofeng Xu
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Wenliang Chen
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ekaterina Turlova
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Nan Dong
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Christopher L F Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yangqingqin Lu
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hanhui Fu
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ranran Shi
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Barszczyk
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dongzi Yang
- Department of Obstetrics and Gynecology, Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Tianru Jin
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Edoardo Mannucci
- Diabetology, Careggi Hospital, University of Florence, Florence, Italy
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Pharmacology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Effect of synthetic cannabinoids on spontaneous neuronal activity: Evaluation using Ca 2+ spiking and multi-electrode arrays. Eur J Pharmacol 2016; 786:148-160. [DOI: 10.1016/j.ejphar.2016.05.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/18/2016] [Accepted: 05/30/2016] [Indexed: 01/22/2023]
|
11
|
Tauskela JS, Aylsworth A, Hewitt M, Brunette E, Blondeau N. Failure and rescue of preconditioning-induced neuroprotection in severe stroke-like insults. Neuropharmacology 2016; 105:533-542. [DOI: 10.1016/j.neuropharm.2016.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 01/30/2023]
|
12
|
Ryanodine receptors contribute to the induction of ischemic tolerance. Brain Res Bull 2016; 122:45-53. [DOI: 10.1016/j.brainresbull.2016.02.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/14/2015] [Accepted: 02/24/2016] [Indexed: 11/21/2022]
|
13
|
Chen X, Liu Y, Zhu J, Lei S, Dong Y, Li L, Jiang B, Tan L, Wu J, Yu S, Zhao Y. GSK-3β downregulates Nrf2 in cultured cortical neurons and in a rat model of cerebral ischemia-reperfusion. Sci Rep 2016; 6:20196. [PMID: 26838164 PMCID: PMC4738318 DOI: 10.1038/srep20196] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 12/23/2015] [Indexed: 12/30/2022] Open
Abstract
The NF-E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway plays a critical role in protecting against oxidative stress in brain ischemia and reperfusion injury. Glycogen synthase kinase 3β (GSK-3β) may play a critical role in regulating Nrf2 in a Kelch-like ECH-associated protein 1 (Keap1)-independent manner. However, the relationship between GSK-3β and Nrf2 in brain ischemia and reperfusion injury is not clear. In this study, we explored the mechanisms through which GSK-3β regulates Nrf2 and Nrf-2/ARE pathways in vitro and in vivo. We used oxygen and glucose deprivation/reoxygenation (OGD/R) in primary cultured cortical neurons and a middle cerebral artery occlusion-reperfusion (MCAO/R) rat model to mimic ischemic insult. In this study, GSK-3β siRNA and inhibitors (SB216763 and LiCl) were used to inhibit GSK-3β in vitro and in vivo. After inhibiting GSK-3β, expression of total and nuclear Nrf2, Nrf2-ARE binding activity, and expression of Nrf2/ARE pathway-driven genes HO-1 and NQO-1 increased. Overexpression of GSK-3β yielded opposite results. These results suggest that GSK-3β downregulates Nrf2 and the Nrf2/ARE pathway in brain ischemia and reperfusion injury. GSK-3β may be an endogenous antioxidant relevant protein, and may represent a new therapeutic target in treatment of ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Yuanling Liu
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Jin Zhu
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Shipeng Lei
- Department of Respiratory Medicine, Jiangjin Center Hospital, 402260, Chongqing, People's Republic of China
| | - Yuan Dong
- Department of Forensic Medicine, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Lingyu Li
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Beibei Jiang
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Li Tan
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Jingxian Wu
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Shanshan Yu
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Institute of Neuroscience, Chongqing Medical University, 400016 Chongqing, People's Republic of China.,Key Laboratory of Neurobiology, Chongqing Medical University, 400016 Chongqing, People's Republic of China
| |
Collapse
|
14
|
Thompson JW, Dawson VL, Perez-Pinzon MA, Dawson TM. Intracellular Signaling. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Kim HJ, Yang JS, Yoon SH. Brief low [Mg(2+)]o-induced Ca(2+) spikes inhibit subsequent prolonged exposure-induced excitotoxicity in cultured rat hippocampal neurons. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 20:101-9. [PMID: 26807029 PMCID: PMC4722183 DOI: 10.4196/kjpp.2016.20.1.101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/23/2015] [Accepted: 11/30/2015] [Indexed: 12/18/2022]
Abstract
Reducing [Mg2+]o to 0.1 mM can evoke repetitive [Ca2+]i spikes and seizure activity, which induces neuronal cell death in a process called excitotoxicity. We examined the issue of whether cultured rat hippocampal neurons preconditioned by a brief exposure to 0.1 mM [Mg2+]o are rendered resistant to excitotoxicity induced by a subsequent prolonged exposure and whether Ca2+ spikes are involved in this process. Preconditioning by an exposure to 0.1 mM [Mg2+]o for 5 min inhibited significantly subsequent 24 h exposure-induced cell death 24 h later (tolerance). Such tolerance was prevented by both the NMDA receptor antagonist D-AP5 and the L-type Ca2+ channel antagonist nimodipine, which blocked 0.1 mM [Mg2+]o-induced [Ca2+]i spikes. The AMPA receptor antagonist NBQX significantly inhibited both the tolerance and the [Ca2+]i spikes. The intracellular Ca2+ chelator BAPTA-AM significantly prevented the tolerance. The nonspecific PKC inhibitor staurosporin inhibited the tolerance without affecting the [Ca2+]i spikes. While Gö6976, a specific inhibitor of PKCα had no effect on the tolerance, both the PKCε translocation inhibitor and the PKCζ pseudosubstrate inhibitor significantly inhibited the tolerance without affecting the [Ca2+]i spikes. Furthermore, JAK-2 inhibitor AG490, MAPK kinase inhibitor PD98059, and CaMKII inhibitor KN-62 inhibited the tolerance, but PI-3 kinase inhibitor LY294,002 did not. The protein synthesis inhibitor cycloheximide significantly inhibited the tolerance. Collectively, these results suggest that low [Mg2+]o preconditioning induced excitotoxic tolerance was directly or indirectly mediated through the [Ca2+]i spike-induced activation of PKCε and PKCξ, JAK-2, MAPK kinase, CaMKII and the de novo synthesis of proteins.
Collapse
Affiliation(s)
- Hee Jung Kim
- Department of Physiology, College of Medicine, Dankook University, Cheonan 31116, Korea
| | - Ji Seon Yang
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Shin Hee Yoon
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.; Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
16
|
Simon RP. Epigenetic modulation of gene expression governs the brain's response to injury. Neurosci Lett 2015; 625:16-9. [PMID: 26739198 DOI: 10.1016/j.neulet.2015.12.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/03/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
Mild stress from ischemia, seizure, hypothermia, or infection can produce a transient neuroprotected state in the brain. In the neuroprotected state, the brain responds differently to a severe stress and sustains less injury. At the genomic level, the response of the neuroprotected brain to a severe stress is characterized by widespread differential regulation of genes with diverse functions. This reprogramming of gene expression observed in the neuroprotected brain in response to a stress is consistent with an epigenetic model of regulation mediated by changes in DNA methylation and histone modification. Here, we summarize our evolving understanding of the molecular basis for endogenous neuroprotection and review recent findings that implicate DNA methylation and protein mediators of histone modification as epigenetic regulators of the brain's response to injury.
Collapse
Affiliation(s)
- Roger P Simon
- Translational Stroke Program, Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA; Grady Memorial Hospital, Atlanta, GA, USA.
| |
Collapse
|
17
|
Klawitter M, Hakozaki M, Kobayashi H, Krupkova O, Quero L, Ospelt C, Gay S, Hausmann O, Liebscher T, Meier U, Sekiguchi M, Konno SI, Boos N, Ferguson SJ, Wuertz K. Expression and regulation of toll-like receptors (TLRs) in human intervertebral disc cells. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2014; 23:1878-91. [PMID: 24997157 DOI: 10.1007/s00586-014-3442-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 06/10/2014] [Accepted: 06/25/2014] [Indexed: 12/15/2022]
Abstract
PURPOSE Although inflammatory processes play an essential role in painful intervertebral disc (IVD) degeneration, the underlying regulatory mechanisms are not well understood. This study was designed to investigate the expression, regulation and importance of specific toll-like receptors (TLRs)--which have been shown to play an essential role e.g. in osteoarthritis--during degenerative disc disease. METHODS The expression of TLRs in human IVDs was measured in isolated cells as well as in normal or degenerated IVD tissue. The role of IL-1β or TNF-α in regulating TLRs (expression/activation) as well as in regulating activity of down-stream pathways (NF-κB) and expression of inflammation-related genes (IL-6, IL-8, HSP60, HSP70, HMGB1) was analyzed. RESULTS Expression of TLR1/2/3/4/5/6/9/10 was detected in isolated human IVD cells, with TLR1/2/4/6 being dependent on the degree of IVD degeneration. Stimulation with IL-1β or TNF-α moderately increased TLR1/TLR4 mRNA expression (TNF-α only), and strongly increased TLR2 mRNA expression (IL-1β/TNF-α), with the latter being confirmed on the protein level. Stimulation with IL-1β, TNF-α or Pam3CSK4 (a TLR2-ligand) stimulated IL-6 and IL-8, which was inhibited by a TLR2 neutralizing antibody for Pam3CSK4; IL-1β and TNF-α caused NF-κB activation. HSP60, HSP70 and HMGB1 did not increase IL-6 or IL-8 and were not regulated by IL-1β/TNF-α. CONCLUSION We provide evidence that several TLRs are expressed in human IVD cells, with TLR2 possibly playing the most crucial role. As TLRs mediate catabolic and inflammatory processes, increased levels of TLRs may lead to aggravated disc degeneration, chronic inflammation and pain development. Especially with the identification of more endogenous TLR ligands, targeting these receptors may hold therapeutic promise.
Collapse
Affiliation(s)
- Marina Klawitter
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Neurons are more vulnerable to oxidative stress than astrocytes, the reasons for which have yet to be fully elucidated. Understanding the cellular and molecular mechanisms which contribute to this enhanced vulnerability is key to efforts aimed at ameliorating neuronal health and resilience to oxidative stress, particularly in the context of neurodegenerative disease, which is characterized by progressive dysfunction and loss of neurons specifically, and in which oxidative stress is considered a central aetiological contributor. Biological factors which may influence neuronal susceptibility to oxidative stress, in normal and neurodegenerative contexts, are reviewed in the present article, with a focus on properties intrinsic to the neuronal cell type and on properties related to neuronal reliance on surrounding astrocytes.
Collapse
|
19
|
Mitochondrial respiratory chain and creatine kinase activities following trauma brain injury in brain of mice preconditioned with N-methyl-d-aspartate. Mol Cell Biochem 2013; 384:129-37. [DOI: 10.1007/s11010-013-1790-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/23/2013] [Indexed: 10/26/2022]
|
20
|
Force spectroscopy measurements show that cortical neurons exposed to excitotoxic agonists stiffen before showing evidence of bleb damage. PLoS One 2013; 8:e73499. [PMID: 24023686 PMCID: PMC3758302 DOI: 10.1371/journal.pone.0073499] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/22/2013] [Indexed: 12/12/2022] Open
Abstract
In ischemic and traumatic brain injury, hyperactivated glutamate (N-methyl-D-aspartic acid, NMDA) and sodium (Nav) channels trigger excitotoxic neuron death. Na+, Ca++ and H2O influx into affected neurons elicits swelling (increased cell volume) and pathological blebbing (disassociation of the plasma membrane’s bilayer from its spectrin-actomyosin matrix). Though usually conflated in injured tissue, cell swelling and blebbing are distinct processes. Around an injury core, salvageable neurons could be mildly swollen without yet having suffered the bleb-type membrane damage that, by rendering channels leaky and pumps dysfunctional, exacerbates the excitotoxic positive feedback spiral. Recognizing when neuronal inflation signifies non-lethal osmotic swelling versus blebbing should further efforts to salvage injury-penumbra neurons. To assess whether the mechanical properties of osmotically-swollen versus excitotoxically-blebbing neurons might be cytomechanically distinguishable, we measured cortical neuron elasticity (gauged via atomic force microscopy (AFM)-based force spectroscopy) upon brief exposure to hypotonicity or to excitotoxic agonists (glutamate and Nav channel activators, NMDA and veratridine). Though unperturbed by solution exchange per se, elasticity increased abruptly with hypotonicity, with NMDA and with veratridine. Neurons then invariably softened towards or below the pre-treatment level, sometimes starting before the washout. The initial channel-mediated stiffening bespeaks an abrupt elevation of hydrostatic pressure linked to NMDA or Nav channel-mediated ion/H2O fluxes, together with increased [Ca++]int-mediated submembrane actomyosin contractility. The subsequent softening to below-control levels is consistent with the onset of a lethal level of bleb damage. These findings indicate that dissection/identification of molecular events during the excitotoxic transition from stiff/swollen to soft/blebbing is warranted and should be feasible.
Collapse
|
21
|
Wu X, Zhao J, Yu S, Chen Y, Wu J, Zhao Y. Sulforaphane protects primary cultures of cortical neurons against injury induced by oxygen-glucose deprivation/reoxygenation via antiapoptosis. Neurosci Bull 2012; 28:509-16. [PMID: 23054633 PMCID: PMC5561925 DOI: 10.1007/s12264-012-1273-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Accepted: 03/14/2012] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE To determine whether sulforaphane (SFN) protects neurons against injury caused by oxygen-glucose deprivation/reoxygenation (OGD/R) and, if so, to investigate the possible mechanisms. METHODS Primary cultures of neurons were prepared from the cerebral cortex of 1-day-old Sprague-Dawley rats. On days 5-6 in vitro, the neurons were exposed to OGD for 1 h, followed by reoxygenation for 24 h. Cells were treated with 0, 0.1, 0.2, 0.5, 1, 2.5, or 5 μmol/L SFN, with or without 10 μmol/L LY294002, a PI3K-specific inhibitor, during OGD/R (a total of 25 h). After 24-h reoxygenation, MTT was used to assess viability and injury was assessed by Hoechst 33258/propidium iodide (PI) staining; immunofluorescence staining and Western blot were performed to detect molecular events associated with apoptosis. RESULTS The MTT assay showed that 1 μmol/L SFN significantly increased viability, and Hoechst 33258/PI staining showed that the numbers of injured neurons were reduced significantly in the SFN group. Furthermore, immunofluorescence staining and Western blot showed that SFN increased Bcl-2 and decreased cleaved caspase-3 levels. Moreover, LY294002 inhibited the phosphorylated-Akt expression evoked by SFN, decreased Bcl-2 expression and increased cleaved caspase-3 expression. CONCLUSION SFN protects neurons against injury from OGD/R and this effect may be partly associated with an antiapoptosis pathway.
Collapse
Affiliation(s)
- Xuemei Wu
- Department of Pathology, Chongqing Medical University, Chongqing, 400016 China
| | - Jing Zhao
- Department of Pathophysiology, Chongqing Medical University, Chongqing, 400016 China
| | - Shanshan Yu
- Department of Pathology, Chongqing Medical University, Chongqing, 400016 China
| | - Yanlin Chen
- Department of Pathology, Chongqing Medical University, Chongqing, 400016 China
| | - Jingxian Wu
- Department of Pathology, Chongqing Medical University, Chongqing, 400016 China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing, 400016 China
| |
Collapse
|
22
|
Lymphocyte cell kinase activation mediates neuroprotection during ischemic preconditioning. J Neurosci 2012; 32:7278-86. [PMID: 22623673 DOI: 10.1523/jneurosci.6273-11.2012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The molecular mechanisms underlying preconditioning (PC), a powerful endogenous neuroprotective phenomenon, remain to be fully elucidated. Once identified, these endogenous mechanisms could be manipulated for therapeutic gain. We investigated whether lymphocyte cell kinase (Lck), a member of the Src kinases family, mediates PC. We used both in vitro primary cortical neurons and in vivo mouse cerebral focal ischemia models of preconditioning, cellular injury, and neuroprotection. Genetically engineered mice deficient in Lck, gene silencing using siRNA, and pharmacological approaches were used. Cortical neurons preconditioned with sublethal exposure to NMDA or oxygen glucose deprivation (OGD) exhibited enhanced Lck kinase activity, and were resistant to injury on subsequent exposure to lethal levels of NMDA or OGD. Lck gene silencing using siRNA abolished tolerance against both stimuli. Lck-/- mice or neurons isolated from Lck-/- mice did not exhibit PC-induced tolerance. An Lck antagonist administered to wild-type mice significantly attenuated the neuroprotective effect of PC in the mouse focal ischemia model. Using pharmacological and gene silencing strategies, we also showed that PKCε is an upstream regulator of Lck, and Fyn is a downstream target of Lck. We have discovered that Lck plays an essential role in PC in both cellular and animal models of stroke. Our data also show that the PKCε-Lck-Fyn axis is a key mediator of PC. These findings provide new opportunities for stroke therapy development.
Collapse
|
23
|
Prohibitin reduces mitochondrial free radical production and protects brain cells from different injury modalities. J Neurosci 2012; 32:583-92. [PMID: 22238093 DOI: 10.1523/jneurosci.2849-11.2012] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Prohibitin is an essential mitochondrial protein that has been implicated in a wide variety of functions in many cell types, but its role in neurons remains unclear. In a proteomic screen of rat brains in which ischemic tolerance was induced by electrical stimulation of the cerebellar fastigial nucleus, we found that prohibitin is upregulated in mitochondria. This observation prompted us to investigate the role of prohibitin in neuronal death and survival. We found that prohibitin is upregulated also in the ischemic tolerance induced by transient ischemia in vivo, or oxygen-glucose deprivation in neuronal cultures. Cell fractionation and electron-microscopic immunolabeling studies demonstrated that prohibitin is localized to neuronal mitochondria. Upregulation of prohibitin in neuronal cultures or hippocampal slices was markedly neuroprotective, whereas prohibitin gene silencing increased neuronal vulnerability, an effect associated with loss of mitochondrial membrane potential and increased mitochondrial production of reactive oxygen species. Prohibitin upregulation was associated with reduced production of reactive oxygen species in mitochondria exposed to the complex I inhibitor rotenone. In addition, prohibitin protected complex I activity from the inhibitory effects of rotenone. These observations, collectively, establish prohibitin as an endogenous neuroprotective protein involved in ischemic tolerance. Prohibitin exerts beneficial effects on neurons by reducing mitochondrial free radical production. The data with complex I activity suggest that prohibitin may stabilize the function of complex I. The protective effect of prohibitin has potential translational relevance in diseases of the nervous system associated with mitochondrial dysfunction and oxidative stress.
Collapse
|
24
|
Bell KFS, Fowler JH, Al-Mubarak B, Horsburgh K, Hardingham GE. Activation of Nrf2-regulated glutathione pathway genes by ischemic preconditioning. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2011; 2011:689524. [PMID: 21904646 PMCID: PMC3166574 DOI: 10.1155/2011/689524] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 04/29/2011] [Indexed: 01/01/2023]
Abstract
Prophylactic pharmacological activation of astrocytic gene expression driven by the transcription factor Nrf2 boosts antioxidant defences and protects against neuronal loss in ischemia and other disease models. However, the role of Nrf2 in mediating endogenous neuroprotective responses is less clear. We recently showed that Nrf2 is activated by mild oxidative stress in both rodent and human astrocytes. Moreover, brief exposure to ischemic conditions was found to activate Nrf2 both in vivo and in vitro, and this was found to contribute to neuroprotective ischemic preconditioning. Here we show that transient ischemic conditions in vitro and in vivo cause an increase in the expression of Nrf2 target genes associated with the glutathione pathway, including those involved in glutathione biosynthesis and cystine uptake. Taken together, these studies indicate that astrocytic Nrf2 may represent an important mediator of endogenous neuroprotective preconditioning pathways.
Collapse
Affiliation(s)
- Karen F. S. Bell
- Centre for Integrative Physiology, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - Jill H. Fowler
- Centre for Cognitive and Neural Systems, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Bashayer Al-Mubarak
- Centre for Integrative Physiology, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | - Karen Horsburgh
- Centre for Cognitive and Neural Systems, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Giles E. Hardingham
- Centre for Integrative Physiology, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
25
|
de Araújo Herculano B, Vandresen-Filho S, Martins WC, Boeck CR, Tasca CI. NMDA preconditioning protects against quinolinic acid-induced seizures via PKA, PI3K and MAPK/ERK signaling pathways. Behav Brain Res 2011; 219:92-7. [DOI: 10.1016/j.bbr.2010.12.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 12/10/2010] [Accepted: 12/13/2010] [Indexed: 10/18/2022]
|
26
|
Saghyan A, LaTorre GN, Keesey R, Sharma A, Mehta V, Rudenko V, Hallas BH, Rafiuddin A, Goldstein B, Friedman LK. Glutamatergic and morphological alterations associated with early life seizure-induced preconditioning in young rats. Eur J Neurosci 2010; 32:1897-911. [DOI: 10.1111/j.1460-9568.2010.07464.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
27
|
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: 1.9] [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
|
28
|
Costa T, Constantino LC, Mendonça BP, Pereira JG, Herculano B, Tasca CI, Boeck CR. N-methyl-D-aspartate preconditioning improves short-term motor deficits outcome after mild traumatic brain injury in mice. J Neurosci Res 2010; 88:1329-37. [PMID: 19998488 DOI: 10.1002/jnr.22300] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Traumatic brain injury (TBI) causes impairment of fine motor functions in humans and nonhuman mammals that often persists for months after the injury occurs. Neuroprotective strategies for prevention of the sequelae of TBI and understanding the molecular mechanisms and cellular pathways are related to the glutamatergic system. It has been suggested that cellular damage subsequent to TBI is mediated by the excitatory neurotransmitters, glutamate and aspartate, through the excessive activation of the N-methyl-D-aspartate (NMDA) receptors. Thus, preconditioning with a low dose of NMDA was used as a strategy for protection against locomotor deficits observed after TBI in mice. Male adult mice CF-1 were preconditioned with NMDA (75 mg/kg) 24 hr before the TBI induction. Under anesthesia with O(2)/N(2)O (33%: 66%) inhalation, the animals were subjected to the experimental model of trauma that occurs by the impact of a 25 g weight on the skull. Sensorimotor gating was evaluated at 1.5, 6, or 24 hr after TBI induction by using footprint and rotarod tests. Cellular damage also was assessed 24 hr after occurrence of cortical trauma. Mice preconditioned with NMDA were protected against all motor deficits revealed by footprint tests, but not those observed in rotarod tasks. Although mice showed motor deficits after TBI, no cellular damage was observed. These data corroborate the hypothesis that glutamatergic excitotoxicity, especially via NMDA receptors, contributes to severity of trauma. They also point to a putative neuroprotective mechanism induced by a sublethal dose of NMDA to improve motor behavioral deficits after TBI.
Collapse
Affiliation(s)
- Tayana Costa
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brasil
| | | | | | | | | | | | | |
Collapse
|
29
|
Dowd WW, Renshaw GMC, Cech JJ, Kültz D. Compensatory proteome adjustments imply tissue-specific structural and metabolic reorganization following episodic hypoxia or anoxia in the epaulette shark (Hemiscyllium ocellatum). Physiol Genomics 2010; 42:93-114. [PMID: 20371547 PMCID: PMC2888556 DOI: 10.1152/physiolgenomics.00176.2009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Accepted: 04/05/2010] [Indexed: 12/31/2022] Open
Abstract
The epaulette shark (Hemiscyllium ocellatum) represents an ancestral vertebrate model of episodic hypoxia and anoxia tolerance at tropical temperatures. We used two-dimensional gel electrophoresis and mass spectrometry-based proteomics approaches, combined with a suite of physiological measures, to characterize this species' responses to 1) one episode of anoxia plus normoxic recovery, 2) one episode of severe hypoxia plus recovery, or 3) two episodes of severe hypoxia plus recovery. We examined these responses in the cerebellum and rectal gland, two tissues with high ATP requirements. Sharks maintained plasma ionic homeostasis following all treatments, and activities of Na(+)/K(+)-ATPase and caspase 3/7 in both tissues were unchanged. Oxygen lack and reoxygenation elicited subtle adjustments in the proteome. Hypoxia led to more extensive proteome responses than anoxia in both tissues. The cerebellum and rectal gland exhibited treatment-specific responses to oxygen limitation consistent with one or more of several strategies: 1) neurotransmitter and receptor downregulation in cerebellum to prevent excitotoxicity, 2) cytoskeletal/membrane reorganization, 3) metabolic reorganization and more efficient intracellular energy shuttling that are more consistent with sustained ATP turnover than with long-term metabolic depression, 4) detoxification of metabolic byproducts and oxidative stress in light of continued metabolic activity, particularly following hypoxia in rectal gland, and 5) activation of prosurvival signaling. We hypothesize that neuronal morphological changes facilitate prolonged protection from excitotoxicity via dendritic spine remodeling in cerebellum (i.e., synaptic structural plasticity). These results recapitulate several highly conserved themes in the anoxia and hypoxia tolerance, preconditioning, and oxidative stress literature in a single system. In addition, several of the identified pathways and proteins suggest potentially novel mechanisms for enhancing anoxia or hypoxia tolerance in vertebrates. Overall, our data show that episodic hypoxic or anoxic exposure and recovery in the epaulette shark amplifies a constitutive suite of compensatory mechanisms that further prepares them for subsequent insults.
Collapse
Affiliation(s)
- W Wesley Dowd
- Department of Animal Science, University of California, Davis, California, USA
| | | | | | | |
Collapse
|
30
|
Friedman LK, Segal M. Early exposure of cultured hippocampal neurons to excitatory amino acids protects from later excitotoxicity. Int J Dev Neurosci 2009; 28:195-205. [DOI: 10.1016/j.ijdevneu.2009.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Revised: 09/10/2009] [Accepted: 11/05/2009] [Indexed: 10/20/2022] Open
Affiliation(s)
- Linda K. Friedman
- Neuroscience DepartmentNew York College of Osteopathic Medicine/New York Institute of TechnologyNorthern BoulevardOld WestburyNY11568United States
| | | |
Collapse
|
31
|
Smith AJ, Tauskela JS, Stone TW, Smith RA. Preconditioning with 4-aminopyridine protects cerebellar granule neurons against excitotoxicity. Brain Res 2009; 1294:165-75. [DOI: 10.1016/j.brainres.2009.07.061] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 07/16/2009] [Accepted: 07/18/2009] [Indexed: 01/23/2023]
|
32
|
Tauskela JS, Brunette E. Neuroprotection against staurosporine by metalloporphyrins independent of antioxidant capability. Neurosci Lett 2009; 466:41-6. [PMID: 19766169 DOI: 10.1016/j.neulet.2009.09.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 08/26/2009] [Accepted: 09/14/2009] [Indexed: 11/29/2022]
Abstract
Metalloporphyrin catalytic antioxidants are remarkably useful in protecting cells and tissues in a wide array of disease models, attributed primarily to functioning as superoxide dismutase (SOD) mimetics or by scavenging other reactive oxygen species (ROS). However, we recently showed that neuroprotection against Ca(2+)-dependent excitotoxic insults did not correlate with antioxidant strength or capability [25], raising the question of whether scavenging of ROS underlies neuroprotection in other types of neuronal injury. The protein kinase inhibitor staurosporine causes neuronal demise primarily by apoptosis. Neuroprotection from staurosporine by a limited number of metalloporphyrin antioxidants has previously been attributed to antioxidant action. In the current study, a wide array of anionic and cationic metalloporphyrins and porphyrins, ranging in antioxidant strength or capability, provided protection against staurosporine in cortical neuron and cerebellar granule neuron (CGN) culture. Neuroprotection did not correlate with antioxidant strength or capability. In CGN but not cortical neuron cultures, NMDA receptor antagonists also prevented neurotoxicity, so metalloporphyrins may also target this secondary mode of death induced by staurosporine. Neuroprotection observed with antioxidant-inactive controls raises the possibility of an additional, or perhaps alternative, mechanism by antioxidant analogs not involving ROS scavenging.
Collapse
Affiliation(s)
- Joseph S Tauskela
- National Research Council, Institute for Biological Sciences, Synaptic Therapies & Devices Group, Montreal Rd. Campus, Ottawa, ON, Canada K1A 0R6.
| | | |
Collapse
|
33
|
Ramadas RA, Wu L, LeVine AM. Surfactant protein A enhances production of secretory leukoprotease inhibitor and protects it from cleavage by matrix metalloproteinases. THE JOURNAL OF IMMUNOLOGY 2009; 182:1560-7. [PMID: 19155504 DOI: 10.4049/jimmunol.182.3.1560] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mice lacking surfactant protein A (SP-A) are susceptible to bacterial infection associated with an excessive inflammatory response in the lung. To determine mechanisms by which SP-A is antiinflammatory in the lung during bacterial infection, SP-A regulation of secretory leukoprotease inhibitor (SLPI), an inhibitor of serine proteases, was assessed. SLPI protein expression and antineutrophil elastase activity were reduced in bronchoalveolar fluid of SP-A(-/-) compared with SP-A(+/+) mice. Intratracheal administration of SP-A to SP-A(-/-) mice enhanced SLPI protein expression and antineutrophil elastase activity in the lung. SLPI mRNA was similar in whole lung and alveolar type II cells; however, it was significantly reduced in alveolar macrophages from SP-A(-/-) compared with SP-A(+/+) mice. In vitro, SP-A enhanced SLPI production by macrophage THP-1 cells but not respiratory epithelial A549 cells. SP-A inhibited LPS induced IkappaB-alpha degradation in THP-1 cells, which was partially reversed with knockdown of SLPI. Matrix metalloproteinase (MMP)-12 cleaved SLPI and incubation with SP-A reduced MMP-12-mediated SLPI cleavage. The collagen-like region of SP-A conferred protection of SLPI against MMP mediated cleavage. SP-A plays an important role in the lung during bacterial infection regulating protease and antiprotease activity.
Collapse
Affiliation(s)
- Ravisankar A Ramadas
- Department of Pediatrics, Division of Critical Care Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | | |
Collapse
|
34
|
Bickler PE, Fahlman CS, Gray J, McKleroy W. Inositol 1,4,5-triphosphate receptors and NAD(P)H mediate Ca2+ signaling required for hypoxic preconditioning of hippocampal neurons. Neuroscience 2009; 160:51-60. [PMID: 19217932 DOI: 10.1016/j.neuroscience.2009.02.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 02/04/2009] [Accepted: 02/06/2009] [Indexed: 01/14/2023]
Abstract
Exposure of neurons to a non-lethal hypoxic stress greatly reduces cell death during subsequent severe ischemia (hypoxic preconditioning, HPC). In organotypic cultures of rat hippocampus, we demonstrate that HPC requires inositol triphosphate (IP3) receptor-dependent Ca2+ release from the endoplasmic reticulum (ER) triggered by increased cytosolic NAD(P)H. Ca2+ chelation with intracellular BAPTA, ER Ca2+ store depletion with thapsigargin, IP3 receptor block with xestospongin, and RNA interference against subtype 1 of the IP3 receptor all blunted the moderate increases in [Ca2+](i) (50-100 nM) required for tolerance induction. Increases in [Ca2+](i) during HPC and neuroprotection following HPC were not prevented with NMDA receptor block or by removing Ca2+ from the bathing medium. Increased NAD(P)H fluorescence in CA1 neurons during hypoxia and demonstration that NADH manipulation increases [Ca2+](i) in an IP3R-dependent manner revealed a primary role of cellular redox state in liberation of Ca2+ from the ER. Blockade of IP3Rs and intracellular Ca2+ chelation prevented phosphorylation of known HPC signaling targets, including MAPK p42/44 (ERK), protein kinase B (Akt) and CREB. We conclude that the endoplasmic reticulum, acting via redox/NADH-dependent intracellular Ca2+ store release, is an important mediator of the neuroprotective response to hypoxic stress.
Collapse
Affiliation(s)
- P E Bickler
- Department of Anesthesia, University of California at San Francisco, 513 Parnassus Avenue, Sciences 255, Box 0542, San Francisco, CA 94143-0542, USA.
| | | | | | | |
Collapse
|
35
|
Tixier E, Leconte C, Touzani O, Roussel S, Petit E, Bernaudin M. Adrenomedullin protects neurons against oxygen glucose deprivation stress in an autocrine and paracrine manner. J Neurochem 2008; 106:1388-403. [DOI: 10.1111/j.1471-4159.2008.05494.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
36
|
Mattson MP. Hormesis and disease resistance: activation of cellular stress response pathways. Hum Exp Toxicol 2008; 27:155-62. [PMID: 18480142 DOI: 10.1177/0960327107083417] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The survival of all organisms depends upon their ability to overcome stressful conditions, an ability that involves adaptive changes in cells and molecules. Findings from studies of animal models and human populations suggest that hormesis (beneficial effects of low levels of stress) is an effective means of protecting against many different diseases including diabetes, cardiovascular disease, cancers and neurodegenerative disorders. Such stress resistance mechanisms can be bolstered by diverse environmental factors including exercise, dietary restriction, cognitive stimulation and exposure to low levels of toxins. Some commonly used vitamins and dietary supplements may also induce beneficial stress responses. Several interrelated cellular signaling molecules are involved in the process of hormesis. Examples include the gases oxygen, carbon monoxide and nitric oxide, the neurotransmitter glutamate, the calcium ion and tumor necrosis factor. In each case low levels of these signaling molecules are beneficial and protect against disease, whereas high levels can cause the dysfunction and/or death of cells. The cellular and molecular mechanisms of hormesis are being revealed and include activation of growth factor signaling pathways, protein chaperones, cell survival genes and enzymes called sirtuins. Knowledge of hormesis mechanisms is leading to novel approaches for preventing and treating a range of human diseases.
Collapse
Affiliation(s)
- Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224, USA.
| |
Collapse
|
37
|
Preconditioning with NMDA protects against toxicity of 3-nitropropionic acid or glutamate in cultured cerebellar granule neurons. Neurosci Lett 2008; 440:294-8. [PMID: 18565656 DOI: 10.1016/j.neulet.2008.05.066] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Revised: 05/09/2008] [Accepted: 05/20/2008] [Indexed: 12/29/2022]
Abstract
A brief sub-lethal ischaemic stimulus has been reported to protect against subsequent ischaemic damage in vivo, and in vitro following periods of hypoxia or oxygen-glucose deprivation (OGD). Preconditioning against neurotoxic stimuli has been linked to N-methyl-d-aspartate (NMDA) receptors, since receptor blockade prevents the protection afforded by OGD, and low doses of NMDA treatment are capable of preconditioning. The current study demonstrated that NMDA preconditioning also protects against 3-nitropropionic acid (3-NPA), a generator of both excitotoxic and oxidative damage, in addition to glutamate. Cerebellar granule neuronal (CGN) cultures prepared from 8-day neonatal Sprague-Dawley rats were maintained for 8 days prior to NMDA stimulation for 6h. At 9 days in vitro (DIV), preconditioned and control cultures were subjected to a toxic insult (1 microM-10 mM glutamate or 1 microM-10 mM 3-NPA). Neuronal viability was assessed by use of a fluorescein diacetate assay. Protection was effective with 100 microM NMDA preconditioning for 6 h against 1-100 microM glutamate, and also against 1-500 microM 3-NPA. The study demonstrates that NMDA preconditioning can be beneficial against excitotoxic treatments, even when these are potentially complicated by associated oxidative damage and metabolic compromise, as is the case for 3-NPA.
Collapse
|
38
|
Kato S, Morimoto SI, Hiramitsu S, Uemura A, Ohtsuki M, Kato Y, Miyagishima K, Mori N, Hishida H. Successful high-dose intravenous immunoglobulin therapy for a patient with fulminant myocarditis. Heart Vessels 2007; 22:48-51. [PMID: 17285446 DOI: 10.1007/s00380-006-0923-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2005] [Accepted: 04/28/2006] [Indexed: 11/25/2022]
Abstract
A 45-year-old man developed fulminant myocarditis for which ventricular assist devices (intra-aortic balloon pumping and percutaneous cardiopulmonary support) were required for hemodynamic support. Echocardiography showed left ventricular akinesis and, since no improvement was noted on the following day, immunoglobulin (70 g/day for 2 days) was added to the therapy. The left ventricular ejection fraction increased to 25% and 40% at 12 and 36 h, respectively, representing a marked improvement in wall motion within a very short period. An endomyocardial biopsy specimen revealed focal lymphomononuclear infiltrate with adjacent myocytolysis, and acute lymphocytic myocarditis was diagnosed. Two days after administration of immunoglobulin, the serum level of interleukin-6 decreased rapidly from 180 to 5.9 pg/ml. In this patient, cardiac function improved immediately after immunoglobulin administration, suggesting the usefulness of this therapy. Three years after the diagnosis the patient is in good health, with steady normal left ventricular ejection fraction. We conclude that there are cases of acute myocarditis in which high-dose intravenous immunoglobulin therapy is effective.
Collapse
Affiliation(s)
- Shigeru Kato
- Division of Cardiology, Department of Internal Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Shimada K, Kishimoto C, Okabe TA, Hattori M, Murayama T, Yokode M, Kita T. Exercise Training Reduces Severity of Atherosclerosis in Apolipoprotein E Knockout Mice via Nitric Oxide. Circ J 2007; 71:1147-51. [PMID: 17587726 DOI: 10.1253/circj.71.1147] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Exercise training may protect against the development of atherosclerosis, although the precise mechanisms are still unknown. The present study assessed the hypothesis that exercise training would reduce the severity of experimental atherosclerosis in apolipoprotein-E (apoE)-deficient mice via nitric oxide (NO). METHODS AND RESULTS ApoE-deficient mice fed a high-fat diet underwent exercise training (30 min swimming) 3 times per week for 8 weeks. The exercise group were also given oral N(G)-nitro-L-arginine methylester (L-NAME; 25 mg x kg (-1) x day(-1)), an inhibitor of NO synthase. Fatty streak plaque lesions developed in ApoE-deficient mice fed the high-fat diet, and were suppressed in the mice that underwent swimming training. In contrast, atherosclerotic lesions were not ameliorated in mice that had exercise training plus oral L-NAME treatment. Immunohistochemical analysis revealed that the expression of endothelial NO increased in mice undergoing exercise compared with the mice that did not exercise, and that the expression was suppressed in the mice having exercise plus oral L-NAME treatment. Differences in lesion area did not correlate with any significant alterations in serum lipid levels. CONCLUSION Exercise training suppressed atherosclerosis via the NO system.
Collapse
Affiliation(s)
- Kana Shimada
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Okabe TA, Kishimoto C, Shimada K, Murayama T, Yokode M, Kita T. Effects of MCI-186 (edaravone), a novel free radical scavenger, upon experimental atherosclerosis in apolipoprotein E-deficient mice. Circ J 2006; 70:1216-9. [PMID: 16936439 DOI: 10.1253/circj.70.1216] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Recent evidence suggests that oxidative stress may play a role in the development of atherosclerosis. MCI-186 (3-methyl-1-phenyl-1-phyrazolin-5-one, edaravone) is a novel free radical scavenger, but it remains unclear whether free radical scavengers would be effective for the prevention of the disease. METHODS AND RESULTS Experimental atherosclerosis was induced in apolipoprotein E-deficient mice fed a high-fat diet containing 0.3% cholesterol. Mice were treated with an intraperitoneal injection of either MCI-186 1 mg/kg per day or MCI-186 10 mg/kg per day on alternate days over 4 weeks. Fatty streak lesion was suppressed by MCI-186 10 mg/kg per day administration, but not by mg/kg per day. Immunohistochemical analysis showed that macrophage and CD4+ T-cell accumulation and oxidative stress overload in the fatty streak lesion were suppressed in mice that received MCI-186 treatment. CONCLUSIONS MCI-186 administration suppressed the development of atherosclerosis, associated with reduced expression of both immune-activated cells and oxidative stress in fatty streak plaques.
Collapse
Affiliation(s)
- Taka-aki Okabe
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Kim HA, Cho ML, Choi HY, Yoon CS, Jhun JY, Oh HJ, Kim HY. The catabolic pathway mediated by Toll-like receptors in human osteoarthritic chondrocytes. ARTHRITIS AND RHEUMATISM 2006; 54:2152-63. [PMID: 16802353 DOI: 10.1002/art.21951] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To examine the catabolic pathways mediated by Toll-like receptor (TLR) ligands in human osteoarthritic (OA) chondrocytes. METHODS The presence of TLRs in OA and non-OA articular cartilage was analyzed by immunohistochemistry. The regulation of TLR messenger RNA (mRNA) by interleukin-1 (IL-1) and tumor necrosis factor alpha (TNFalpha) was analyzed by reverse transcription-polymerase chain reaction. For stimulation of TLR-2 and TLR-4, chondrocytes were treated with Staphylococcus aureus peptidoglycan and lipopolysaccharides (LPS), respectively. Production of matrix metalloproteinases (MMPs) 1, 3, and 13 and prostaglandin E2 (PGE2) was evaluated by enzyme-linked immunosorbent assay. Production of nitric oxide (NO) was analyzed by the Griess reaction. Regulation of cyclooxygenase 2 protein and phosphorylation of MAPKs (p38, ERK, and JNK) were evaluated by Western blotting or solid-phase kinase assay. NF-kappaB activation was evaluated by electrophoretic mobility shift assay. RESULTS Expression of TLRs 2 and 4 was up-regulated in lesional areas of OA cartilage. Treatment with IL-1, TNFalpha, peptidoglycan, and LPS all significantly up-regulated TLR-2 mRNA expression in cultured chondrocytes. Production of MMPs 1, 3, and 13 and of NO and PGE2 was significantly increased after treating chondrocytes with either of the TLR ligands. Prolonged culture of cartilage explants with TLR ligands also led to a significant increase in the release of proteoglycan and type II collagen degradation product. Treatment with TLR ligands led to phosphorylation of all 3 MAPKs and activation of NF-kappaB. CONCLUSION We found that TLRs are increased in OA cartilage lesions. TLR-2 and TLR-4 ligands strongly induce catabolic responses in chondrocytes. Modulation of TLR-mediated signaling as a therapeutic strategy would require detailed elucidation of the signaling pathways involved.
Collapse
Affiliation(s)
- Hyun Ah Kim
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, 896, Pyongchon-dong, Dongan-gu, Anyang, Kyunggi-do 431-070, Republic of Korea.
| | | | | | | | | | | | | |
Collapse
|
42
|
Tauskela JS, Brunette E, Hewitt M, Mealing G, Morley P. Competing approaches to excitotoxic neuroprotection by inert and catalytic antioxidant porphyrins. Neurosci Lett 2006; 401:236-41. [PMID: 16631306 DOI: 10.1016/j.neulet.2006.03.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Revised: 03/03/2006] [Accepted: 03/15/2006] [Indexed: 10/24/2022]
Abstract
The goal of this study was to determine if novel porphyrins protect cultured cortical neurons from excitotoxic NMDA exposure or oxygen-glucose deprivation (OGD), which model key aspects of cerebral ischemia. Porphyrins were chosen based on conventional and unconventional criteria. Metalloporphyrin catalytic antioxidants possessing a redox-sensitive metal core can exhibit potent and wide-ranging catalytic antioxidant abilities, which are conventionally believed to underlie neuroprotection. We report here that a recent-generation potent peroxynitrite decomposition catalyst, FP-15, protected a majority of neurons against OGD and NMDA toxicity, without suppressing NMDA-mediated intracellular Ca2+ (Cai2+) elevations or whole-cell currents. We have previously shown that neuroprotection against OGD and NMDA toxicity correlated with an ability to suppress neurotoxic Cai2+ elevations and not antioxidant ability. We now evaluate if this unconventional mechanism extends to inert metal-free porphyrins. Neuron cultures were completely protected against OGD and NMDA toxicity by H2-meso-tetrakis(3-benzoic acid)porphyrin (H2-TBAP(3)) or H2-meso-tetrakis(4-sulfonatophenyl)porphyrin (H2-TPPS(4)), although only H2-TPPS(4) suppressed (completely) NMDA-induced Cai2+ rises. H2-meso-tetrakis(3,3'-benzoic acid)porphyrin (H2-TBAP(3,3')) or H2-meso-tetrakis(N-methylpyridynium-4-yl)porphyrin (H2-TM-PyP(4)) provided at least partial protection against OGD and NMDA toxicity and partially suppressed NMDA-induced Cai2+ elevations. Despite the complexity of Ca2+-independent and -dependent based mechanisms, the inventory of porphyrins demonstrating neuroprotection in ischemia-relevant insults is now expanded to include FP-15 and inert metal-free compounds, although with no apparent advantage gained by using FP-15.
Collapse
Affiliation(s)
- Joseph Stephen Tauskela
- National Research Council, Institute for Biological Sciences, Synaptic Pathophysiology Group, Montreal Road Campus, Building M-54, Ottawa, Ontario, Canada K1A 0R6.
| | | | | | | | | |
Collapse
|
43
|
Raval AP, Bramlett H, Perez-Pinzon MA. Estrogen preconditioning protects the hippocampal CA1 against ischemia. Neuroscience 2006; 141:1721-30. [PMID: 16777351 DOI: 10.1016/j.neuroscience.2006.05.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Revised: 04/30/2006] [Accepted: 05/09/2006] [Indexed: 01/06/2023]
Abstract
Estrogen is neuroprotective against ischemia in both in vivo and in vitro injury models. Because of the promising preclinical data on neuroprotection, the Women's Estrogen for Stroke Trial was initiated. The outcomes from this trial were, however, unsuccessful and questions emerged about the safety of chronic estrogen treatment in women. In contrast to the chronic estrogen treatment strategy, the present study aims to investigate: (1) the neuroprotective efficacy of single estrogen pretreatment/preconditioning; and (2) the existence of a similarity between estrogen- and ischemic preconditioning-induced neuroprotection against cerebral ischemia. The efficacy of estrogen was tested in an in vitro model of cerebral ischemia using hippocampal organotypic slice culture system. The hippocampal organotypic slice cultures were generated from female neonatal (9-11 days old) Sprague-Dawley rats. The slices were exposed to estradiol-17beta (0.5, 1, 5 nM) for various durations (1, 2 or 4 h) 48 h prior to ischemia (40 min of oxygen-glucose deprivation). For ischemic preconditioning, slices were exposed to sublethal oxygen-glucose deprivation (15 min), 48 h prior to lethal oxygen-glucose deprivation. Quantification of cell death in hippocampal CA1 region was conducted by using propidium iodide fluorescence staining technique. Results demonstrated that estrogen preconditioning significantly protects the hippocampal CA1 region against ischemia (P<0.001) and mimicked ischemic preconditioning-induced neuroprotection. The propidium iodide fluorescence values of estrogen preconditioning, ischemic preconditioning and ischemia groups were 21+/-2 (mean+/-S.E.M.) (1 nM; 2 h; n=15), 18+/-2 (5 nM; 4 h; n=12), 32+/-3 (n=8), 65+/-3 (n=27), respectively. Further, estrogen preconditioning initiated a calcium-mediated signaling pathway leading to protection of CA1 neurons against ischemia. Future investigations in estrogen preconditioning may suggest new estrogen regimens that avoid potential side effects of chronic estrogen treatment for stroke patients.
Collapse
Affiliation(s)
- A P Raval
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program (D4-5), P.O. Box 016960, University of Miami School of Medicine, Miami, FL 33101, USA.
| | | | | |
Collapse
|
44
|
Haddad JJ. N-methyl-D-aspartate (NMDA) and the regulation of mitogen-activated protein kinase (MAPK) signaling pathways: a revolving neurochemical axis for therapeutic intervention? Prog Neurobiol 2006; 77:252-82. [PMID: 16343729 DOI: 10.1016/j.pneurobio.2005.10.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2003] [Revised: 12/10/2004] [Accepted: 10/27/2005] [Indexed: 12/30/2022]
Abstract
Excitatory synaptic transmission in the central nervous system (CNS) is mediated by the release of glutamate from presynaptic terminals onto postsynaptic channels gated by N-methyl-D-aspartate (NMDA) and non-NMDA (AMPA and KA) receptors. Extracellular signals control diverse neuronal functions and are responsible for mediating activity-dependent changes in synaptic strength and neuronal survival. Influx of extracellular calcium ([Ca(2+)](e)) through the NMDA receptor (NMDAR) is required for neuronal activity to change the strength of many synapses. At the molecular level, the NMDAR interacts with signaling modules, which, like the mitogen-activated protein kinase (MAPK) superfamily, transduce excitatory signals across neurons. Recent burgeoning evidence points to the fact that MAPKs play a crucial role in regulating the neurochemistry of NMDARs, their physiologic and biochemical/biophysical properties, and their potential role in pathophysiology. It is the purpose of this review to discuss: (i) the MAPKs and their role in a plethora of cellular functions; (ii) the role of MAPKs in regulating the biochemistry and physiology of NMDA receptors; (iii) the kinetics of MAPK-NMDA interactions and their biologic and neurochemical properties; (iv) how cellular signaling pathways, related cofactors and intracellular conditions affect NMDA-MAPK interactions and (v) the role of NMDA-MAPK pathways in pathophysiology and the evolution of disease conditions. Given the versatility of the NMDA-MAPK interactions, the NMDA-MAPK axis will likely form a neurochemical target for therapeutic interventions.
Collapse
Affiliation(s)
- John J Haddad
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Lebanon.
| |
Collapse
|
45
|
Okabe TA, Kishimoto C, Shimada K, Murayama T, Yokode M, Kita T. Effects of late administration of immunoglobulin on experimental atherosclerosis in apolipoprotein E-deficient mice. Circ J 2006; 69:1543-6. [PMID: 16308506 DOI: 10.1253/circj.69.1543] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although immunoglobulin treatment, beginning simultaneously with the initiation of atherosclerosis, suppresses experimental atherosclerosis in apolipoprotein E-deficient mice, it remains unclear whether the treatment at a subsequent stage of atherosclerosis would be effective. METHODS AND RESULTS Experimental atherosclerosis was induced in mice fed a high-fat diet containing 0.3% cholesterol. After confirming the presence of atherosclerotic lesions at 11 weeks, the mice were treated with an intraperitoneal injection of either intact type of immunoglobulin or F(ab')2 fragments of immunoglobulin (both, 1 g.kg-1.day-1) on alternate days over 4 weeks. Fatty streak lesion was suppressed by intact immunoglobulin administration, but not by F(ab')2 fragments of immunoglobulin. Immunohistochemical analysis showed that macrophage and CD4+ T-cell accumulation in the fatty streak lesion was suppressed in mice that received intact immunoglobulin but not in those that received F(ab')2 fragments. CONCLUSIONS Immunoglobulin treatment, even at a later stage of atherosclerosis, suppresses the development of lesions associated with the reduced expression of immune-activated cells in fatty streak plaques, demonstrating the benefits of immunoglobulin therapy for prevention of atherosclerosis.
Collapse
Affiliation(s)
- Taka-aki Okabe
- Department of Cadiovascular Medicine, Graduate School of Medicine, Kyoto University, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Hoshi A, Nakahara T, Kayama H, Yamamoto T. Ischemic tolerance in chemical preconditioning: Possible role of astrocytic glutamine synthetase buffering glutamate-mediated neurotoxicity. J Neurosci Res 2006; 84:130-41. [PMID: 16688719 DOI: 10.1002/jnr.20869] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glutamine synthetase (GS), localized to astrocyte is a key enzyme in the glutamate-glutamine pathway in the brain. 3-Nitropropionic acid (3-NPA) is an irreversible inhibitor of succinate dehydrogenase in the tricarboxylic-acid cycle, and provides ischemic tolerance to the brain. So far, there have been no reports on the relationship of astrocytic GS and ischemic tolerance by chemical preconditioning. In order to test the hypothesis that astrocytes serve a pivotal role in 3-NPA-induced chemical preconditioning, we have investigated the temporal profile of GS expression in astrocyte parallel with those of glial fibrillary acidic protein and heat-shock protein 70 (HSP70). In our rat model of permanent focal ischemia, preconditioning with 3-NPA singnificantly reduced the subsequent neurological deficits and infarct volume within 24-72 hours after treatment. Immunohistochemically, protoplasmic astrocytes in the cortex and striatum were activated in terms of upregulation of GS and more abundant protoplasmic processes with 3-NPA preconditioning, however, HSP70 expression could not be induced. Thus, the activation of astrocytes and upregulation of GS play an important role in 3-NPA-induced preconditioning but HSP70 does not. In view of glutamate being imposed on the cerebral ischemic damage, the astrocytic GS may contribute to 3-NPA-induced ischemic tolerance.
Collapse
Affiliation(s)
- Akihiko Hoshi
- Department of Neurology, Fukushima Medical University, Fukushima, Japan
| | | | | | | |
Collapse
|
47
|
Jiang SX, Lertvorachon J, Hou ST, Konishi Y, Webster J, Mealing G, Brunette E, Tauskela J, Preston E. Chlortetracycline and Demeclocycline Inhibit Calpains and Protect Mouse Neurons against Glutamate Toxicity and Cerebral Ischemia. J Biol Chem 2005; 280:33811-8. [PMID: 16091365 DOI: 10.1074/jbc.m503113200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Minocycline is a potent neuroprotective tetracycline in animal models of cerebral ischemia. We examined the protective properties of chlortetracycline (CTC) and demeclocycline (DMC) and showed that these two tetracyclines were also potent neuroprotective against glutamate-induced neuronal death in vitro and cerebral ischemia in vivo. However, CTC and DMC appeared to confer neuroprotection through a unique mechanism compared with minocycline. Rather than inhibiting microglial activation and caspase, CTC and DMC suppressed calpain activities. In addition, CTC and DMC only weakly antagonized N-methyl-D-aspartate (NMDA) receptor activities causing 16 and 14%, respectively, inhibition of NMDA-induced whole cell currents and partially blocked NMDA-induced Ca2+ influx, commonly regarded as the major trigger of neuronal death. In vitro and in vivo experiments demonstrated that the two compounds selectively inhibited the activities of calpain I and II activated following glutamate treatment and cerebral ischemia. In contrast, minocycline did not significantly inhibit calpain activity. Taken together, these results suggested that CTC and DMC provide neuroprotection through suppression of a rise in intracellular Ca2+ and inhibition of calpains.
Collapse
Affiliation(s)
- Susan X Jiang
- Neurophysiology Group, National Research Council Institute for Biological Sciences, National Research Council Canada, Ottawa, Ontario K1A 0R6, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Chen ZH, Yoshida Y, Saito Y, Niki E. Adaptation to hydrogen peroxide enhances PC12 cell tolerance against oxidative damage. Neurosci Lett 2005; 383:256-9. [PMID: 15878237 DOI: 10.1016/j.neulet.2005.04.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2005] [Revised: 04/01/2005] [Accepted: 04/05/2005] [Indexed: 02/08/2023]
Abstract
The adaptive responses to H2O2 and the resulting protective effect against oxidative stress have been investigated using PC12 cells. Pretreatment of sublethal doses of H2O2 significantly protected PC12 cells against the cytotoxicity induced by lethal H2O2. The endogenous antioxidant defense systems, catalase and glutathione peroxidase, were enhanced quickly by the pretreatment of low doses of H2O2. This pretreatment also exerted protective effect against the oxidative insults induced by 6-hydroxydopamine and paraquat, but not against alkyl peroxyl radicals. Our results, taken together, suggest that the stimulation by low dose of H2O2 enriches the cellular antioxidant defense systems, thereby enhancing cell tolerance against the forthcoming oxidative insults induced by H2O2 and related hydroxyl radicals.
Collapse
Affiliation(s)
- Zhi-Hua Chen
- Human Stress Signal Research Center (HSSRC), National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31, Ikeda, Osaka 563-8577, Japan.
| | | | | | | |
Collapse
|
49
|
Tauskela JS, Brunette E, O'Reilly N, Mealing G, Comas T, Gendron TF, Monette R, Morley P. An alternative Ca2+‐dependent mechanism of neuroprotection by the metalloporphyrin class of superoxide dismutase mimetics. FASEB J 2005; 19:1734-6. [PMID: 16081500 DOI: 10.1096/fj.05-3795fje] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This study challenges the conventional view that metalloporphyrins protect cultured cortical neurons in models of cerebral ischemia by acting as intracellular catalytic antioxidants [superoxide dismutase (SOD) mimetics]. High SOD-active Mn(III)porphyrins meso-substituted with N,N'-dimethylimidazolium or N-alkylpyridinium groups did not protect neurons against oxygen-glucose deprivation (OGD), although lower SOD-active and -inactive para isomers protected against N-methyl-D-aspartate (NMDA) exposure. Mn(III)meso-tetrakis(4-benzoic acid)porphyrin (Mn(III)TBAP), as well as SOD-inactive metalloTBAPs and other phenyl ring- or beta-substituted metalloporphyrins that contained redox-insensitive metals, protected cultures against OGD and NMDA neurotoxicity. Crucially, neuroprotective metalloporphyrins suppressed OGD- or NMDA-induced rises in intracellular Ca2+ concentration in the same general rank order as observed for neuroprotection. Results from paraquat toxicity, intracellular fluorescence quenching, electrophysiology, mitochondrial Ca2+, and spontaneous synaptic activity experiments suggest a model in which metalloporphyrins, acting at the plasma membrane, protect neurons against OGD by suppressing postsynaptic NMDA receptor-mediated Ca2+ rises, thereby indirectly preventing accumulation of neurotoxic mitochondrial Ca2+ levels. Though neuroprotective in a manner not originally intended, SOD-inactive metalloporphyrins may represent promising therapeutic agents in diseases such as cerebral ischemia, in which Ca2+ toxicity is implicated. Conventional syntheses aimed at improving the catalytic antioxidant capability and/or intracellular access of metalloporphyrins may not yield improved efficacy in some disease models.
Collapse
Affiliation(s)
- Joseph S Tauskela
- National Research Council, Institute for Biological Sciences, Synaptic Pathophysiology Group, Ottawa, ON, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Furuichi T, Liu W, Shi H, Miyake M, Liu KJ. Generation of hydrogen peroxide during brief oxygen-glucose deprivation induces preconditioning neuronal protection in primary cultured neurons. J Neurosci Res 2005; 79:816-24. [PMID: 15668910 DOI: 10.1002/jnr.20402] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although reactive oxygen species (ROS) have been implicated in ischemic preconditioning (IPC)-induced neuronal protection, several key questions concerning ROS remain to be elucidated. The purpose of this study is to obtain direct evidence for the formation of specific ROS species generated by IPC, and to determine the specific species that is responsible for the observed neuronal protection. Primary cultured cortex neurons from rat embryos were preconditioned with 10 min of oxygen-glucose deprivation (OGD), which increased the intracellular levels of superoxide and hydrogen peroxide. This preconditioning markedly induced neuronal protection against 2-hr OGD stimuli. Preconditioning with exogenous ROS by the administration of xanthine/xanthine oxidase (X/XO), or hydrogen peroxide was also found to induce IPC-like neuronal protection. Administration of hydrogen peroxide scavengers, such as catalase, glutathione, or the thiol reductant N-(2-mercaptopriopionyl)-glycine, all reduced the increase in the intracellular hydrogen peroxide levels, which effectively eliminated IPC- or exogenous ROS-induced neuronal protection. In contrast, administration of the membrane-permeable superoxide dismutase mimic Mn(III)tetrakis(1-methyl-4-pyridyl)porphyrin pentachloride was able to block the increase of intracellular superoxide levels during IPC, but did not abolish either IPC- or exogenous X/XO preconditioning-induced neuronal protection. These findings strongly suggest that IPC enhances the generation of superoxide, which is then converted to hydrogen peroxide, and that hydrogen peroxide is likely the main trigger involved in the mechanism of IPC-induced neuronal protection.
Collapse
Affiliation(s)
- Takamitsu Furuichi
- College of Pharmacy, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | | | | | | | | |
Collapse
|