1
|
Bao L, Liu Y, Jia Q, Chu S, Jiang H, He S. Argon neuroprotection in ischemic stroke and its underlying mechanism. Brain Res Bull 2024; 212:110964. [PMID: 38670471 DOI: 10.1016/j.brainresbull.2024.110964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/04/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024]
Abstract
Ischemic stroke (IS), primarily caused by cerebrovascular obstruction, results in severe neurological deficits and has emerged as a leading cause of death and disability worldwide. Recently, there has been increasing exploration of the neuroprotective properties of the inert gas argon. Argon has exhibited impressive neuroprotection in many in vivo and ex vivo experiments without signs of adverse effects, coupled with the advantages of being inexpensive and easily available. However, the efficient administration strategy and underlying mechanisms of neuroprotection by argon in IS are still unclear. This review summarizes current research on the neuroprotective effects of argon in IS with the goal to provide effective guidance for argon application and to elucidate the potential mechanisms of argon neuroprotection. Early and appropriate argon administration at as high a concentration as possible offers favorable neuroprotection in IS. Argon inhalation has been shown to provide some long-term protection benefits. Argon provides the anti-oxidative stress, anti-inflammatory and anti-apoptotic cytoprotective effects mainly around Toll-like receptor 2/4 (TLR2/4), mediated by extracellular signal-regulated kinase 1/2 (ERK1/2), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), nuclear factor kappa-B (NF-ĸB) and B-cell leukemia/lymphoma 2 (Bcl-2). Therefore, argon holds significant promise as a novel clinical neuroprotective gas agent for ischemic stroke after further researches to identify the optimal application strategy and elucidate the underlying mechanism.
Collapse
Affiliation(s)
- Li Bao
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Yongxin Liu
- Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Qi Jia
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Sihao Chu
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Han Jiang
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Medical College of Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Shuang He
- Department of Stroke Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China.
| |
Collapse
|
2
|
Nascimento AA, Pereira-Figueiredo D, Borges-Martins VP, Kubrusly RC, Calaza KC. GABAergic system and chloride cotransporters as potential therapeutic targets to mitigate cell death in ischemia. J Neurosci Res 2024; 102:e25355. [PMID: 38808645 DOI: 10.1002/jnr.25355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 04/17/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
Gamma aminobutyric acid (GABA) is a critical inhibitory neurotransmitter in the central nervous system that plays a vital role in modulating neuronal excitability. Dysregulation of GABAergic signaling, particularly involving the cotransporters NKCC1 and KCC2, has been implicated in various pathologies, including epilepsy, schizophrenia, autism spectrum disorder, Down syndrome, and ischemia. NKCC1 facilitates chloride influx, whereas KCC2 mediates chloride efflux via potassium gradient. Altered expression and function of these cotransporters have been associated with excitotoxicity, inflammation, and cellular death in ischemic events characterized by reduced cerebral blood flow, leading to compromised tissue metabolism and subsequent cell death. NKCC1 inhibition has emerged as a potential therapeutic approach to attenuate intracellular chloride accumulation and mitigate neuronal damage during ischemic events. Similarly, targeting KCC2, which regulates chloride efflux, holds promise for improving outcomes and reducing neuronal damage under ischemic conditions. This review emphasizes the critical roles of GABA, NKCC1, and KCC2 in ischemic pathologies and their potential as therapeutic targets. Inhibiting or modulating the activity of these cotransporters represents a promising strategy for reducing neuronal damage, preventing excitotoxicity, and improving neurological outcomes following ischemic events. Furthermore, exploring the interactions between natural compounds and NKCC1/KCC2 provides additional avenues for potential therapeutic interventions for ischemic injury.
Collapse
Affiliation(s)
- A A Nascimento
- Neurobiology of the Retina Laboratory, Department of Neurobiology and Graduate Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - D Pereira-Figueiredo
- Graduate Program in Biomedical Sciences (Physiology and Pharmacology), Fluminense Federal University, Niterói, Brazil
| | - V P Borges-Martins
- Laboratory of Neuropharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - R C Kubrusly
- Laboratory of Neuropharmacology, Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - K C Calaza
- Neurobiology of the Retina Laboratory, Department of Neurobiology and Graduate Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
- Graduate Program in Biomedical Sciences (Physiology and Pharmacology), Fluminense Federal University, Niterói, Brazil
| |
Collapse
|
3
|
Lan J, Wang J, Wang S, Wang J, Huang S, Wang Y, Ma Y. The Activation of GABA AR Alleviated Cerebral Ischemic Injury via the Suppression of Oxidative Stress, Autophagy, and Apoptosis Pathways. Antioxidants (Basel) 2024; 13:194. [PMID: 38397792 PMCID: PMC10886019 DOI: 10.3390/antiox13020194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Ischemic stroke is a devastating disease leading to neurologic impairment. Compounding the issue is the very limited array of available interventions. The activation of a γ-aminobutyric acid (GABA) type A receptor (GABAAR) has been reported to produce neuroprotective properties during cerebral ischemia, but its mechanism of action is not yet fully understood. Here, in a rat model of photochemically induced cerebral ischemia, we found that muscimol, a GABAAR agonist, modulated GABAergic signaling, ameliorated anxiety-like behaviors, and attenuated neuronal damage in rats suffering cerebral ischemia. Moreover, GABAAR activation improved brain antioxidant levels, reducing the accumulation of oxidative products, which was closely associated with the NO/NOS pathway. Notably, the inhibition of autophagy markedly relieved the neuronal insult caused by cerebral ischemia. We further established an oxygen-glucose deprivation (OGD)-induced PC12 cell injury model. Both in vivo and in vitro experiments demonstrated that GABAAR activation obviously suppressed autophagy by regulating the AMPK-mTOR pathway. Additionally, GABAAR activation inhibited apoptosis through inhibiting the Bax/Bcl-2 pathway. These data suggest that GABAAR activation exerts neuroprotective effects during cerebral ischemia through improving oxidative stress and inhibiting autophagy and apoptosis. Our findings indicate that GABAAR serves as a target for treating cerebral ischemia and highlight the GABAAR-mediated autophagy signaling pathway.
Collapse
Affiliation(s)
- Jing Lan
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jiaqi Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Shujing Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jia Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Sijuan Huang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yazhou Wang
- Department of Neurobiology, School of Basic Medicine, The Fourth Military Medical University, Xi’an 710032, China
| | - Yunfei Ma
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
4
|
Benke D, Bhat MA, Hleihil M. GABAB Receptors: Molecular Organization, Function, and Alternative Drug Development by Targeting Protein-Protein Interactions. THE RECEPTORS 2024:3-39. [DOI: 10.1007/978-3-031-67148-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
5
|
Christidis P, Vij A, Petousis S, Ghaemmaghami J, Shah BV, Koutroulis I, Kratimenos P. Neuroprotective effect of Src kinase in hypoxia-ischemia: A systematic review. Front Neurosci 2022; 16:1049655. [PMID: 36507364 PMCID: PMC9730728 DOI: 10.3389/fnins.2022.1049655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2022] Open
Abstract
Background Hypoxic-ischemic encephalopathy (HIE) is a major cause of neonatal morbidity and mortality worldwide. While the application of therapeutic hypothermia has improved neurodevelopmental outcomes for some survivors of HIE, this lone treatment option is only available to a subset of affected neonates. Src kinase, an enzyme central to the apoptotic cascade, is a potential pharmacologic target to preserve typical brain development after HIE. Here, we present evidence of the neuroprotective effects of targeting Src kinase in preclinical models of HIE. Methods We performed a comprehensive literature search using the National Library of Medicine's MEDLINE database to compile studies examining the impact of Src kinase regulation on neurodevelopment in animal models. Each eligible study was assessed for bias. Results Twenty studies met the inclusion criteria, and most studies had an intermediate risk for bias. Together, these studies showed that targeting Src kinase resulted in a neuroprotective effect as assessed by neuropathology, enzymatic activity, and neurobehavioral outcomes. Conclusion Src kinase is an effective neuroprotective target in the setting of acute hypoxic injury. Src kinase inhibition triggers multiple signaling pathways of the sub-membranous focal adhesions and the nucleus, resulting in modulation of calcium signaling and prevention of cell death. Despite the significant heterogeneity of the research studies that we examined, the available evidence can serve as proof-of-concept for further studies on this promising therapeutic strategy.
Collapse
Affiliation(s)
- Panagiotis Christidis
- Laboratory of Physiology, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Abhya Vij
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Stamatios Petousis
- 2nd Department of Obstetrics and Gynecology, “Hippokrateion” General Hospital of Thessaloniki, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Javid Ghaemmaghami
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC, United States
| | - Bhairav V. Shah
- Division of Pediatric Surgery, Department of Pediatrics, School of Medicine, Prisma Health Children's Hospital-Midlands, University of South Carolina, Columbia, SC, United States
| | - Ioannis Koutroulis
- Department of Pediatrics, Division of Emergency Medicine, Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Panagiotis Kratimenos
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC, United States,Division of Neonatology, Department of Pediatrics, Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC, United States,*Correspondence: Panagiotis Kratimenos
| |
Collapse
|
6
|
Balakrishnan K, Hleihil M, Bhat MA, Ganley RP, Vaas M, Klohs J, Zeilhofer HU, Benke D. Targeting the interaction of GABA B receptors with CaMKII with an interfering peptide restores receptor expression after cerebral ischemia and inhibits progressive neuronal death in mouse brain cells and slices. Brain Pathol 2022; 33:e13099. [PMID: 35698024 PMCID: PMC9836377 DOI: 10.1111/bpa.13099] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/29/2022] [Indexed: 01/21/2023] Open
Abstract
Cerebral ischemia is the leading cause for long-term disability and mortality in adults due to massive neuronal death. Currently, there is no pharmacological treatment available to limit progressive neuronal death after stroke. A major mechanism causing ischemia-induced neuronal death is the excessive release of glutamate and the associated overexcitation of neurons (excitotoxicity). Normally, GABAB receptors control neuronal excitability in the brain via prolonged inhibition. However, excitotoxic conditions rapidly downregulate GABAB receptors via a CaMKII-mediated mechanism and thereby diminish adequate inhibition that could counteract neuronal overexcitation and neuronal death. To prevent the deleterious downregulation of GABAB receptors, we developed a cell-penetrating synthetic peptide (R1-Pep) that inhibits the interaction of GABAB receptors with CaMKII. Administration of this peptide to cultured cortical neurons exposed to excitotoxic conditions restored cell surface expression and function of GABAB receptors. R1-Pep did not affect CaMKII expression or activity but prevented its T286 autophosphorylation that renders it autonomously and persistently active. Moreover, R1-Pep counteracted the aberrant downregulation of G protein-coupled inwardly rectifying K+ channels and the upregulation of N-type voltage-gated Ca2+ channels, the main effectors of GABAB receptors. The restoration of GABAB receptors activated the Akt survival pathway and inhibited excitotoxic neuronal death with a wide time window in cultured neurons. Restoration of GABAB receptors and neuroprotective activity of R1-Pep was verified by using brain slices prepared from mice after middle cerebral artery occlusion (MCAO). Treatment with R1-Pep restored normal GABAB receptor expression and GABA receptor-mediated K+ channel currents. This reduced MCAO-induced neuronal excitability and inhibited neuronal death. These results support the hypothesis that restoration of GABAB receptor expression under excitatory conditions provides neuroprotection and might be the basis for the development of a selective intervention to inhibit progressive neuronal death after ischemic stroke.
Collapse
Affiliation(s)
- Karthik Balakrishnan
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland,Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland,Present address:
Dewpoint Therapeutics GMBHDresdenGermany
| | - Mohammad Hleihil
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland,Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland
| | - Musadiq A. Bhat
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland
| | - Robert P. Ganley
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland
| | - Markus Vaas
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland,Present address:
Clinical Trial Center ZurichUniversity Hospital of ZurichZurichSwitzerland
| | - Jan Klohs
- Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland,Institute for Biomedical Engineering, University of Zurich and ETH ZurichZurichSwitzerland
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland,Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland,Drug Discovery Network ZurichZurichSwitzerland,Institute of Pharmaceutical Sciences, ETH ZurichZurichSwitzerland
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University of ZurichZurichSwitzerland,Neuroscience Center ZurichUniversity of Zurich and ETH ZurichZurichSwitzerland,Drug Discovery Network ZurichZurichSwitzerland
| |
Collapse
|
7
|
Bhat MA, Esmaeili A, Neumann E, Balakrishnan K, Benke D. Targeting the Interaction of GABA B Receptors With CHOP After an Ischemic Insult Restores Receptor Expression and Inhibits Progressive Neuronal Death. Front Pharmacol 2022; 13:870861. [PMID: 35422706 PMCID: PMC9002115 DOI: 10.3389/fphar.2022.870861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 01/01/2023] Open
Abstract
GABAB receptors control neuronal excitability via slow and prolonged inhibition in the central nervous system. One important function of GABAB receptors under physiological condition is to prevent neurons from shifting into an overexcitation state which can lead to excitotoxic death. However, under ischemic conditions, GABAB receptors are downregulated, fostering over-excitation and excitotoxicity. One mechanism downregulating GABAB receptors is mediated via the interaction with the endoplasmic reticulum (ER) stress-induced transcription factor CHOP. In this study, we investigated the hypothesis that preventing the interaction of CHOP with GABAB receptors after an ischemic insult restores normal expression of GABAB receptors and reduces neuronal death. For this, we designed an interfering peptide (R2-Pep) that restored the CHOP-induced downregulation of cell surface GABAB receptors in cultured cortical neurons subjected to oxygen and glucose deprivation (OGD). Administration of R2-Pep after OGD restored normal cell surface expression of GABAB receptors as well as GABAB receptor-mediated inhibition. As a result, R2-Pep reduced enhanced neuronal activity and inhibited progressive neuronal death in OGD stressed cultures. Thus, targeting diseases relevant protein-protein interactions might be a promising strategy for developing highly specific novel therapeutics.
Collapse
Affiliation(s)
- Musadiq A Bhat
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Abolghasem Esmaeili
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Elena Neumann
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Karthik Balakrishnan
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Drug Discovery Network Zurich (DDNZ), Zurich, Switzerland
| |
Collapse
|
8
|
Hleihil M, Vaas M, Bhat MA, Balakrishnan K, Benke D. Sustained Baclofen-Induced Activation of GABA B Receptors After Cerebral Ischemia Restores Receptor Expression and Function and Limits Progressing Loss of Neurons. Front Mol Neurosci 2021; 14:726133. [PMID: 34539344 PMCID: PMC8440977 DOI: 10.3389/fnmol.2021.726133] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/12/2021] [Indexed: 11/13/2022] Open
Abstract
One important function of GABAB receptors is the control of neuronal activity to prevent overexcitation and thereby excitotoxic death, which is a hallmark of cerebral ischemia. Consequently, sustained activation of GABAB receptors with the selective agonist baclofen provides neuroprotection in in vitro and in vivo models of cerebral ischemia. However, excitotoxic conditions severely downregulate the receptors, which would compromise the neuroprotective effectiveness of baclofen. On the other hand, recent work suggests that sustained activation of GABAB receptors stabilizes receptor expression. Therefore, we addressed the question whether sustained activation of GABAB receptors reduces downregulation of the receptor under excitotoxic conditions and thereby preserves GABAB receptor-mediated inhibition. In cultured neurons subjected to oxygen and glucose deprivation (OGD), to mimic cerebral ischemia, GABAB receptors were severely downregulated. Treatment of the cultures with baclofen after OGD restored GABAB receptor expression and reduced loss of neurons. Restoration of GABAB receptors was due to enhanced fast recycling of the receptors, which reduced OGD-induced sorting of the receptors to lysosomal degradation. Utilizing the middle cerebral artery occlusion (MCAO) mouse model of cerebral ischemia, we verified the severe downregulation of GABAB receptors in the affected cortex and a partial restoration of the receptors after systemic injection of baclofen. Restored receptor expression recovered GABAB receptor-mediated currents, normalized the enhanced neuronal excitability observed after MCAO and limited progressive loss of neurons. These results suggest that baclofen-induced restoration of GABAB receptors provides the basis for the neuroprotective activity of baclofen after an ischemic insult. Since GABAB receptors regulate multiple beneficial pathways, they are promising targets for a neuroprotective strategy in acute cerebral ischemia.
Collapse
Affiliation(s)
- Mohammad Hleihil
- Institute of Pharmacology and Toxicology, University Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich, University of Zurich, Zurich, Switzerland
| | - Markus Vaas
- Institute of Pharmacology and Toxicology, University Zurich, Zurich, Switzerland
| | - Musadiq A Bhat
- Institute of Pharmacology and Toxicology, University Zurich, Zurich, Switzerland
| | - Karthik Balakrishnan
- Institute of Pharmacology and Toxicology, University Zurich, Zurich, Switzerland
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Engin A, Engin AB. N-Methyl-D-Aspartate Receptor Signaling-Protein Kinases Crosstalk in Cerebral Ischemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:259-283. [PMID: 33539019 DOI: 10.1007/978-3-030-49844-3_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Although stroke is very often the cause of death worldwide, the burden of ischemic and hemorrhagic stroke varies between regions and over time regarding differences in prognosis, prevalence of risk factors, and treatment strategies. Excitotoxicity, oxidative stress, dysfunction of the blood-brain barrier, neuroinflammation, and lysosomal membrane permeabilization, sequentially lead to the progressive death of neurons. In this process, protein kinases-related checkpoints tightly regulate N-methyl-D-aspartate (NMDA) receptor signaling pathways. One of the major hallmarks of cerebral ischemia is excitotoxicity, characterized by overactivation of glutamate receptors leading to intracellular Ca2+ overload and ultimately neuronal death. Thus, reduced expression of postsynaptic density-95 protein and increased protein S-nitrosylation in neurons is responsible for neuronal vulnerability in cerebral ischemia. In this chapter death-associated protein kinases, cyclin-dependent kinase 5, endoplasmic reticulum stress-induced protein kinases, hyperhomocysteinemia-related NMDA receptor overactivation, ephrin-B-dependent amplification of NMDA-evoked neuronal excitotoxicity and lysosomocentric hypothesis have been discussed.Consequently, ample evidences have demonstrated that enhancing extrasynaptic NMDA receptor activity triggers cell death after stroke. In this context, considering the dual roles of NMDA receptors in both promoting neuronal survival and mediating neuronal damage, selective augmentation of NR2A-containing NMDA receptor activation in the presence of NR2B antagonist may constitute a promising therapy for stroke.
Collapse
Affiliation(s)
- Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| |
Collapse
|
10
|
The Effects of GABAergic System under Cerebral Ischemia: Spotlight on Cognitive Function. Neural Plast 2020; 2020:8856722. [PMID: 33061952 PMCID: PMC7539123 DOI: 10.1155/2020/8856722] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/23/2020] [Indexed: 12/31/2022] Open
Abstract
In this review, we present evidence about the changes of the GABAergic system on the hippocampus under the ischemic environment, which may be an underlying mechanism to the ischemia-induced cognitive deficit. GABAergic system, in contrast to the glutamatergic system, is considered to play an inhibitory effect on the central nervous system over the past several decades. It has received widespread attention in the area of schizophrenia and epilepsy. The GABAergic system has a significant effect in promoting neural development and formation of local neural circuits of the brain, which is the structural basis of cognitive function. There have been a number of reviews describing changes in the GABAergic system in cerebral ischemia in recent years. However, no study has investigated the changes in the system in the hippocampus during cerebral ischemic injury, which results in cognitive impairment, particularly at the chronic ischemic stage and the late phase of ischemia. We present a review of the changes of the GABAergic system in the hippocampus under ischemia, including GABA interneurons, extracellular GABA neurotransmitter, and GABA receptors. Several studies are also listed correlating amelioration of cognitive impairment by regulating the GABAergic system in the hippocampus damaged under ischemia. Furthermore, exogenous cell transplantation, which improves cognition by modulating the GABAergic system, will also be described in this review to bring new insight and strategy on solving cognitive deficits caused by cerebral ischemia.
Collapse
|
11
|
Vinnakota C, Govindpani K, Tate WP, Peppercorn K, Anekal PV, Waldvogel HJ, Faull RLM, Kwakowsky A. An 5 GABAA Receptor Inverse Agonist, 5IA, Attenuates Amyloid Beta-Induced Neuronal Death in Mouse Hippocampal Cultures. Int J Mol Sci 2020; 21:ijms21093284. [PMID: 32384683 PMCID: PMC7247548 DOI: 10.3390/ijms21093284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder for which no cognition-restoring therapies exist. Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the brain. Increasing evidence suggests a remodeling of the GABAergic system in AD, which might represent an important therapeutic target. An inverse agonist of α5 subunit-containing GABAA receptors (α5GABAARs), 3-(5-Methylisoxazol-3-yl)-6-[(1-methyl-1,2,3-triazol-4-yl)methyloxy]-1,2,4-triazolo[3–a]phthalazine (α5IA) has cognition-enhancing properties. This study aimed to characterize the effects of α5IA on amyloid beta (Aβ1–42)-induced molecular and cellular changes. Mouse primary hippocampal cultures were exposed to either Aβ1-42 alone, or α5IA alone, α5IA with Aβ1–42 or vehicle alone, and changes in cell viability and mRNA expression of several GABAergic signaling components were assessed. Treatment with 100 nM of α5IA reduced Aβ1–42-induced cell loss by 23.8% (p < 0.0001) after 6 h and by 17.3% after 5 days of treatment (p < 0.0001). Furthermore, we observed an Aβ1-42-induced increase in ambient GABA levels, as well as upregulated mRNA expression of the GABAAR α2,α5,β2/3 subunits and the GABABR R1 and R2 subunits. Such changes in GABARs expression could potentially disrupt inhibitory neurotransmission and normal network activity. Treatment with α5IA restored Aβ1-42-induced changes in the expression of α5GABAARs. In summary, this compound might hold neuroprotective potential and represent a new therapeutic avenue for AD.
Collapse
Affiliation(s)
- Chitra Vinnakota
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Karan Govindpani
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Warren Perry Tate
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (W.P.T.); (K.P.)
| | - Katie Peppercorn
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (W.P.T.); (K.P.)
| | - Praju Vikas Anekal
- Biomedical Imaging Research Unit, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand;
| | - Henry John Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Richard Lewis Maxwell Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
- Correspondence: ; Tel.: +64-9923-9346
| |
Collapse
|
12
|
The Roles of GABA in Ischemia-Reperfusion Injury in the Central Nervous System and Peripheral Organs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4028394. [PMID: 31814874 PMCID: PMC6878816 DOI: 10.1155/2019/4028394] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/27/2019] [Accepted: 10/18/2019] [Indexed: 12/30/2022]
Abstract
Ischemia-reperfusion (I/R) injury is a common pathological process, which may lead to dysfunctions and failures of multiple organs. A flawless medical way of endogenous therapeutic target can illuminate accurate clinical applications. γ-Aminobutyric acid (GABA) has been known as a marker in I/R injury of the central nervous system (mainly in the brain) for a long time, and it may play a vital role in the occurrence of I/R injury. It has been observed that throughout cerebral I/R, levels, syntheses, releases, metabolisms, receptors, and transmissions of GABA undergo complex pathological variations. Scientists have investigated the GABAergic enhancers for attenuating cerebral I/R injury; however, discussions on existing problems and mechanisms of available drugs were seldom carried out so far. Therefore, this review would summarize the process of pathological variations in the GABA system under cerebral I/R injury and will cover corresponding probable issues and mechanisms in using GABA-related drugs to illuminate the concern about clinical illness for accurately preventing cerebral I/R injury. In addition, the study will summarize the increasing GABA signals that can prevent I/R injuries occurring in peripheral organs, and the roles of GABA were also discussed correspondingly.
Collapse
|
13
|
Keilhoff G, Nguyen Thi TM, Esser T, Ebmeyer U. Relative Resilience of Cerebellar Purkinje Cells in a Cardiac Arrest/Resuscitation Rat Model. Neurocrit Care 2019; 32:775-789. [DOI: 10.1007/s12028-019-00799-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
14
|
Liu P, Qin D, Huang X, Chen H, Ye W, Lin X, Su J. Neurotoxicity of sodium salicylate to the spiral ganglion neurons: GABA A receptor regulates NMDA receptor by Fyn-dependent phosphorylation. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2019; 205:469-479. [PMID: 31020389 DOI: 10.1007/s00359-019-01339-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 11/25/2022]
Abstract
The purpose of this study was to observe the regulatory effects of GABAA (γ-aminobutyric acid A) receptor on the N-methyl-D-aspartate (NMDA) receptor during excitotoxicity in spiral ganglion neurons in the rat cochlea induced by sodium salicylate (SS). Western blot illustrated SS decreased the expression of NMDA receptor 2B subunit (NR2B) surface protein through affecting GABAA receptor, but the total protein content did not significantly change. Y1472 and S1480 are important phosphorylation sites in NR2B, SS downregulated the Fyn-dependent phosphorylation of Y1472 in a manner not related to the CK2 (Casein Kinase 2) dependent phosphorylation of S1480, thus regulating the surface distribution and internalization of NMDA receptor through GABAA receptor. These results suggest that the modified pattern of dynamic balance between excitation and inhibition by coactivation of the GABAA receptor can attenuate the excitatory NMDA receptor under the action of SS, via inhibiting the Fyn-dependent phosphorylation of Y1472.
Collapse
Affiliation(s)
- Peiqiang Liu
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Danxue Qin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xi Huang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Otolaryngology-Head and Neck Surgery, Wuhan No. 1 Hospital, Wuhan, 430022, Hubei, China
| | - Huiying Chen
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wenhua Ye
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoyu Lin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiping Su
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
15
|
Jafarian M, Modarres Mousavi SM, Alipour F, Aligholi H, Noorbakhsh F, Ghadipasha M, Gharehdaghi J, Kellinghaus C, Kovac S, Khaleghi Ghadiri M, Meuth SG, Speckmann EJ, Stummer W, Gorji A. Cell injury and receptor expression in the epileptic human amygdala. Neurobiol Dis 2019; 124:416-427. [DOI: 10.1016/j.nbd.2018.12.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 12/04/2018] [Accepted: 12/22/2018] [Indexed: 02/06/2023] Open
|
16
|
Fan YF, Guan SY, Luo L, Li YJ, Yang L, Zhou XX, Guo GD, Zhao MG, Yang Q, Liu G. Tetrahydroxystilbene glucoside relieves the chronic inflammatory pain by inhibiting neuronal apoptosis, microglia activation, and GluN2B overexpression in anterior cingulate cortex. Mol Pain 2018; 14:1744806918814367. [PMID: 30380983 PMCID: PMC6259074 DOI: 10.1177/1744806918814367] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tetrahydroxystilbene glucoside (THSG) is one of the active ingredients of Polygonum multiflorum. It has been shown to exert a variety of pharmacological effects, including antioxidant, anti-aging, and anti-atherosclerosis. Because of its prominent anti-inflammatory effect, we explored whether THSG had analgesic effect. In this study, we used a model of chronic inflammatory pain caused by injecting complete Freund's adjuvant into the hind paw of mice. We found THSG relieved swelling and pain in the hind paw of mice on a dose-dependent manner. In the anterior cingulate cortex, THSG suppressed the upregulation of GluN2B-containing N-methyl-D-aspartate receptors and the downregulation of GluN2A-containing N-methyl-D-aspartate receptors caused by chronic inflammation. In addition, THSG increased Bcl-2 and decreased Bax and Caspase-3 expression by protecting neuronal survival. Furthermore, THSG inhibited the phosphorylation of p38 and the increase of nuclear factor κB (NF-κB) and tumor necrosis factor α (TNF-α). Immunohistochemical staining revealed that THSG blocked the activation of microglia and reduced the release of proinflammatory cytokines TNF-α, interleukin 1β (IL-1β), and interleukin 6 (IL-6). In conclusion, this study demonstrated that THSG had a certain effect on alleviating complete Freund's adjuvant-induced chronic inflammatory pain.
Collapse
Affiliation(s)
- Yong-Fei Fan
- 1 Department of Orthopedics, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Shao-Yu Guan
- 2 Department of Pharmacy, Precision Pharmacy and Drug Development Center, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China.,3 Department of Nature Medicine, School of Pharmacy, Air Force Medical University, Xi'an, China
| | - Li Luo
- 2 Department of Pharmacy, Precision Pharmacy and Drug Development Center, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Yan-Jiao Li
- 2 Department of Pharmacy, Precision Pharmacy and Drug Development Center, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China.,3 Department of Nature Medicine, School of Pharmacy, Air Force Medical University, Xi'an, China
| | - Le Yang
- 2 Department of Pharmacy, Precision Pharmacy and Drug Development Center, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Xuan-Xuan Zhou
- 3 Department of Nature Medicine, School of Pharmacy, Air Force Medical University, Xi'an, China
| | - Guo-Dong Guo
- 1 Department of Orthopedics, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ming-Gao Zhao
- 2 Department of Pharmacy, Precision Pharmacy and Drug Development Center, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Qi Yang
- 2 Department of Pharmacy, Precision Pharmacy and Drug Development Center, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Gang Liu
- 1 Department of Orthopedics, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
17
|
Li K, Zhou H, Zhan L, Shi Z, Sun W, Liu D, Liu L, Liang D, Tan Y, Xu W, Xu E. Hypoxic Preconditioning Maintains GLT-1 Against Transient Global Cerebral Ischemia Through Upregulating Cx43 and Inhibiting c-Src. Front Mol Neurosci 2018; 11:344. [PMID: 30323740 PMCID: PMC6172853 DOI: 10.3389/fnmol.2018.00344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 09/03/2018] [Indexed: 01/06/2023] Open
Abstract
Transient global cerebral ischemia (tGCI) causes excessive release of glutamate from neurons. Astrocytic glutamate transporter-1 (GLT-1) and glutamine synthetase (GS) together play a predominant role in maintaining glutamate at normal extracellular concentrations. Though our previous studies reported the alleviation of tGCI-induced neuronal death by hypoxic preconditioning (HPC) in hippocampal Cornu Ammonis 1 (CA1) of adult rats, the underlying mechanism has not yet been fully elaborated. In this study, we aimed to investigate the roles of GLT-1 and GS in the neuroprotection mediated by HPC against tGCI and to ascertain whether these roles can be regulated by connexin 43 (Cx43) and cellular-Src (c-Src) activity. We found that HPC decreased the level of extracellular glutamate in CA1 after tGCI via maintenance of GLT-1 expression and GS activity. Inhibition of GLT-1 expression with dihydrokainate (DHK) or inhibition of GS activity with methionine sulfoximine (MSO) abolished the neuroprotection induced by HPC. Also, HPC markedly upregulated Cx43 and inhibited p-c-Src expression in CA1 after tGCI, whereas inhibition of Cx43 with Gap26 dramatically reversed this effect. Furthermore, inhibition of p-c-Src with 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo (3, 4-d) pyrimidine (PP2) decreased c-Src activity, increased protein levels of GLT-1 and Cx43, enhanced GS activity, and thus reduced extracellular glutamate level in CA1 after tGCI. Collectively, our data demonstrated that reduced extracellular glutamate induced by HPC against tGCI through preventing the reduction of GLT-1 expression and maintaining GS activity in hippocampal CA1, which was mediated by upregulating Cx43 expression and inhibiting c-Src activity.
Collapse
Affiliation(s)
- Kongping Li
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Huarong Zhou
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Lixuan Zhan
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Zhe Shi
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Weiwen Sun
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Dandan Liu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Liu Liu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Donghai Liang
- Department of Environmental Health Sciences, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Yafu Tan
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China.,Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wensheng Xu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - En Xu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| |
Collapse
|
18
|
Abstract
Stroke, also known as “cerebrovascular accident”, is an acute cerebrovascular disease that is caused by a sudden rupture of blood vessels in the brain or obstruction of the blood supply by blockage of blood vessels, thus including hemorrhagic and ischemic strokes. The incidence of ischemic stroke is higher than that of hemorrhagic stroke, and accounts for 80% of the total number of strokes. However, the mortality rate of hemorrhagic stroke is relatively high. Internal carotid artery and vertebral artery occlusion and stenosis can cause ischemic stroke, and especially males over 40 years of age are at a high risk of morbidity. According to the survey, stroke in urban and rural areas has become the first cause of death in China. It is also the leading cause of disability in Chinese adults. In a word, stroke is characterized by high morbidity, high mortality and high disability rates. Studies have shown that many noble gases have the neuroprotective effects. For example, xenon has been extensively studied in various animal models of neurological injury including stroke, hypoxic-ischemic encephalopathy. Compared to xenon, Argon, as a noble gas, is abundant, cheap and widely applicable, and has been also demonstrated to be neuroprotective in many research studies. In a variety of models, ranging from oxygen-glucose deprivation in cell culture to complex models of mid-cerebral artery occlusion, subarachnoid hemorrhage or retinal ischemia-reperfusion injury in animals. Argon administration after individual injury demonstrated favorable effects, particularly increased cell survival and even improved neuronal function. Therefore the neuroprotective effects of argon may be of possible clinical use for opening a potential therapeutic window in stroke. It is important to illuminate the mechanisms of argon in nerve function and to explore the best use of this gas in stroke treatment.
Collapse
Affiliation(s)
- Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhu-Wei Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jin-Quan Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
19
|
Zimmermann J, Perry A, Breakspear M, Schirner M, Sachdev P, Wen W, Kochan NA, Mapstone M, Ritter P, McIntosh AR, Solodkin A. Differentiation of Alzheimer's disease based on local and global parameters in personalized Virtual Brain models. NEUROIMAGE-CLINICAL 2018; 19:240-251. [PMID: 30035018 PMCID: PMC6051478 DOI: 10.1016/j.nicl.2018.04.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 04/05/2018] [Accepted: 04/14/2018] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) is marked by cognitive dysfunction emerging from neuropathological processes impacting brain function. AD affects brain dynamics at the local level, such as changes in the balance of inhibitory and excitatory neuronal populations, as well as long-range changes to the global network. Individual differences in these changes as they relate to behaviour are poorly understood. Here, we use a multi-scale neurophysiological model, “The Virtual Brain (TVB)”, based on empirical multi-modal neuroimaging data, to study how local and global dynamics correlate with individual differences in cognition. In particular, we modeled individual resting-state functional activity of 124 individuals across the behavioural spectrum from healthy aging, to amnesic Mild Cognitive Impairment (MCI), to AD. The model parameters required to accurately simulate empirical functional brain imaging data correlated significantly with cognition, and exceeded the predictive capacity of empirical connectomes. Modeled local and global dynamics correlate with individual cognition in Alzheimer's. Proof of concept of The Virtual Brain to characterize individual dynamics Brain-behaviour relations depend on the network modeled (whole brain or limbic). Model parameters predict cognition better than metrics of neuroimaging data.
Collapse
Affiliation(s)
- J Zimmermann
- Baycrest Health Sciences, Rotman Research Institute, 3560 Bathurst St, Toronto, Ontario M6A 2E1, Canada.
| | - A Perry
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Program of Mental Health Research, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia; Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany
| | - M Breakspear
- Program of Mental Health Research, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia; Metro North Mental Health Service, Royal Brisbane and Women's Hospital, Herston, QLD 4029, Australia
| | - M Schirner
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Dept. of Neurology, Chariteplatz 1, Berlin 13353, Germany; Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - P Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - W Wen
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - N A Kochan
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - M Mapstone
- UC Irvine Health School of Medicine, Irvine Hall, 1001 Health Sciences Road, Irvine, CA 92697-3950, USA
| | - P Ritter
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Dept. of Neurology, Chariteplatz 1, Berlin 13353, Germany; Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - A R McIntosh
- Baycrest Health Sciences, Rotman Research Institute, 3560 Bathurst St, Toronto, Ontario M6A 2E1, Canada
| | - A Solodkin
- UC Irvine Health School of Medicine, Irvine Hall, 1001 Health Sciences Road, Irvine, CA 92697-3950, USA
| |
Collapse
|
20
|
Amantea D, Bagetta G. Excitatory and inhibitory amino acid neurotransmitters in stroke: from neurotoxicity to ischemic tolerance. Curr Opin Pharmacol 2017; 35:111-119. [DOI: 10.1016/j.coph.2017.07.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 12/22/2022]
|
21
|
Sun Y, Zhan L, Cheng X, Zhang L, Hu J, Gao Z. The Regulation of GluN2A by Endogenous and Exogenous Regulators in the Central Nervous System. Cell Mol Neurobiol 2017; 37:389-403. [PMID: 27255970 PMCID: PMC11482088 DOI: 10.1007/s10571-016-0388-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 05/25/2016] [Indexed: 12/25/2022]
Abstract
The NMDA receptor is the most widely studied ionotropic glutamate receptor, and it is central to many physiological and pathophysiological processes in the central nervous system. GluN2A is one of the two main types of GluN2 NMDA receptor subunits in the forebrain. The proper activity of GluN2A is important to brain function, as the abnormal regulation of GluN2A may induce some neuropsychiatric disorders. This review will examine the regulation of GluN2A by endogenous and exogenous regulators in the central nervous system.
Collapse
Affiliation(s)
- Yongjun Sun
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Liying Zhan
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China
| | - Xiaokun Cheng
- North China Pharmaceutical Group New Drug Research and Development Co., Ltd, Shijiazhuang, 050015, People's Republic of China
| | - Linan Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Jie Hu
- School of Nursing, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Zibin Gao
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China.
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China.
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Shijiazhuang, 050018, People's Republic of China.
| |
Collapse
|
22
|
Sun Y, Cheng X, Hu J, Gao Z. The Role of GluN2A in Cerebral Ischemia: Promoting Neuron Death and Survival in the Early Stage and Thereafter. Mol Neurobiol 2017; 55:1208-1216. [DOI: 10.1007/s12035-017-0395-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/09/2017] [Indexed: 01/10/2023]
|
23
|
Öz P, Saybaşılı H. In vitro detection of oxygen and glucose deprivation-induced neurodegeneration and pharmacological neuroprotection based on hippocampal stratum pyramidale width. Neurosci Lett 2016; 636:196-204. [PMID: 27845243 DOI: 10.1016/j.neulet.2016.11.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/23/2016] [Accepted: 11/10/2016] [Indexed: 12/30/2022]
Abstract
Ischemia is one of the most prominent risk factors of neurodegenerative diseases such as Alzheimer's disease. The effects of oxygen and glucose depletion in hippocampal tissue due to ischemia can be mimicked in vitro using the oxygen and glucose deprivation (OGD) model. In this study, we applied OGD on acute rat hippocampal slices in order to design an elementary yet quantitative histological technique that compares the neuroprotective effects of (l)-carnitine to known neuroprotectors, such as the N-methyl-d-aspartate (NMDA) receptor antagonist memantine and the gamma-aminobutyric acid (GABA)-B receptor agonist baclofen. The level of neurodegeneration and the efficiency of pharmacological applications were estimated via stratum pyramidale width measurements in CA1 and CA3 regions of Nissl-stained 200-μm thick hippocampal slices. We demonstrated that (l)-carnitine is an effective pharmacological target against the neurodegeneration induced by in vitro ischemia in a narrow range of concentrations. Even though the effect of chemical neuroprotection was significant, full recovery was not achieved in the dose interval of 5-100μM. In addition to chemical applications, hypothermia was used as a physical neuroprotection against ischemia-related neurodegeneration. Our results showed that incubation of slices for 60min at 4°C provided the same level of neuroprotection as the most effective doses of memantine, baclofen, and (l)-carnitine.
Collapse
Affiliation(s)
- Pınar Öz
- Neuropsychopharmacology Application and Research Center, Üsküdar University Central Campus, Altunizade Mah. Haluk Türksoy Sk. No:14 34662, Istanbul, Turkey; Institute of Biomedical Engineering, Boğaziçi Üniversity Kandilli Campus, Kandilli Mah., 34684 Istanbul, Turkey
| | - Hale Saybaşılı
- Institute of Biomedical Engineering, Boğaziçi Üniversity Kandilli Campus, Kandilli Mah., 34684 Istanbul, Turkey.
| |
Collapse
|
24
|
Duan ZZ, Zhang F, Li FY, Luan YF, Guo P, Li YH, Liu Y, Qi SH. Protease activated receptor 1 (PAR1) enhances Src-mediated tyrosine phosphorylation of NMDA receptor in intracerebral hemorrhage (ICH). Sci Rep 2016; 6:29246. [PMID: 27385592 PMCID: PMC4935874 DOI: 10.1038/srep29246] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/14/2016] [Indexed: 12/20/2022] Open
Abstract
It has been demonstrated that Src could modulate NMDA receptor, and PAR1 could also affect NMDAR signaling. However, whether PAR1 could regulate NMDAR through Src under ICH has not yet been investigated. In this study, we demonstrated the role of Src-PSD95-GluN2A signaling cascades in rat ICH model and in vitro thrombin challenged model. Using the PAR1 agonist SFLLR, antagonist RLLFS and Src inhibitor PP2, electrophysiological analysis showed that PAR1 regulated NMDA-induced whole-cell currents (INMDA) though Src in primary cultured neurons. Both in vivo and in vitro results showed the elevated phosphorylation of tyrosine in Src and GluN2A and enhanced interaction of the Src-PSD95-GluN2A under model conditions. Treatment with the PAR1 antagonist RLLFS, AS-PSD95 (Antisense oligonucleotide against PSD95) and Src inhibitor PP2 inhibited the interaction among Src-PSD95-GluN2A, and p-Src, p-GluN2A. Co-application of SFLLR and AS-PSD95, PP2, or MK801 (NMDAR inhibitor) abolished the effect of SF. In conclusion, our results demonstrated that activated thrombin receptor PAR1 induced Src activation, enhanced the interaction among Src-PSD95-GluN2A signaling modules, and up-regulated GluN2A phosphorylation after ICH injury. Elucidation of such signaling cascades would possibly provide novel targets for ICH treatment.
Collapse
Affiliation(s)
- Zhen-Zhen Duan
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Feng Zhang
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Feng-Ying Li
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Yi-Fei Luan
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Peng Guo
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Yi-Hang Li
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Yong Liu
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Su-Hua Qi
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| |
Collapse
|
25
|
Li M, Qiu L, Wang L, Wang W, Xin L, Li Y, Liu Z, Song L. The inhibitory role of γ-aminobutyric acid (GABA) on immunomodulation of Pacific oyster Crassostrea gigas. FISH & SHELLFISH IMMUNOLOGY 2016; 52:16-22. [PMID: 26975413 DOI: 10.1016/j.fsi.2016.03.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 03/03/2016] [Accepted: 03/10/2016] [Indexed: 05/15/2023]
Abstract
γ-aminobutyric acid (GABA) is an inhibitory neurotransmitter to suppress the immune-mediated pro-inflammatory reactions, and it has been used in the treatment of many inflammation-related diseases in vertebrates, while its immunomodulatory role in invertebrates has never been reported. In the present study, GABA was found to exist in the hemolymph of Pacific oyster Crassostrea gigas, and its concentration decreased slightly from 8.00 ± 0.37 μmol L(-1) at normal condition to 7.73 ± 0.15 μmol L(-1) at 6 h after LPS stimulation, and then increased to 9.34 ± 0.15 μmol L(-1), 8.86 ± 0.68 μmol L(-1) at 12 h and 48 h, respectively. After LPS stimulation, the mRNA expressions of pro-inflammatory cytokines (CgIL-17 and CgTNF) and immune effectors (CgSOD and CgBPI), and the protein expression of NOS increased significantly, and these increased trends were remarkably inhibited by GABA stimulation. At the same time, the phagocytosis rate and apoptosis rate of immunocytes also increased obviously after LPS stimulation, whereas the increase was repressed with the addition of GABA. The results collectively demonstrated that GABA was an indispensable inhibitory agent for both humoral and cellular immune response, which mainly functioned at the late phase of immune response to avoid the excess immune reactions and maintain the immune homeostasis.
Collapse
Affiliation(s)
- Meijia Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Lingling Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian 116023, China.
| | - Weilin Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lusheng Xin
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiqun Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaoqun Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linsheng Song
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
26
|
Ding Y, Xie L, Chang CQ, Chen ZM, Ai H. Activation of γ-aminobutyric Acid (A) Receptor Protects Hippocampus from Intense Exercise-induced Synapses Damage and Apoptosis in Rats. Chin Med J (Engl) 2016; 128:2330-9. [PMID: 26315081 PMCID: PMC4733790 DOI: 10.4103/0366-6999.163392] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Our previous study has confirmed that one bout of exhaustion (Ex) can cause hippocampus neurocyte damage, excessive apoptosis, and dysfunction. Its initial reason is intracellular calcium overload in hippocampus triggered by N-methyl-D-aspartic acid receptor (NMDAR) over-activation. NMDAR activation can be suppressed by γ-aminobutyric acid (A) receptor (GABAAR). Whether GABAAR can prevent intense exercise-induced hippocampus apoptosis, damage, or dysfunction will be studied in this study. METHODS According to dose test, rats were randomly divided into control (Con), Ex, muscimol (MUS, 0.1 mg/kg) and bicuculline (BIC, 0.5 mg/kg) groups, then all rats underwent once swimming Ex except ones in Con group only underwent training. Intracellular free calcium concentration ([Ca2+]i) was measured by Fura-2-acetoxymethyl ester; glial librillary acidic protein (GFAP) and synaptophysin (SYP) immunofluorescence were also performed; apoptosis were displayed by dUTP nick end labeling (TUNEL) stain; endoplasmic reticulum stress-induced apoptosis pathway was detected by Western blotting analysis; Morris water maze was used to detect learning ability and spatial memory. RESULTS The appropriate dose was 0.1 mg/kg for MUS and 0.5 mg/kg for BIC. Ex group showed significantly increased [Ca2+]i and astrogliosis; TUNEL positive cells and levels of GFAP, B cell lymphoma-2 (Bcl-2) associated X protein (Bax), caspase-3, caspase-12 cleavage, CCAAT/enhancer binding protein homologous protein (CHOP), and p-Jun amino-terminal kinase (p-JNK) in Ex group also raised significantly compared to Con group, while SYP, synapse plasticity, and Bcl-2 levels in Ex group were significantly lower than those in Con group. These indexes were back to normal in MUS group. BIC group had the highest levels of [Ca2+]i, astrogliosis, TUNEL positive cell, GFAP, Bax, caspase-3, caspase-12 cleavage, CHOP, and p-JNK, it also gained the lowest SYP, synapse plasticity, and Bcl-2 levels among all groups. Water maze test showed that Ex group had longer escape latency (EL) and less quadrant dwell time than Con group; all indexes between MUS and Con groups had no significant differences; BIC had the longest EL and least quadrant dwell time among all groups. CONCLUSIONS Activation of GABAA R could prevent intense exercise-induced synapses damage, excessive apoptosis, and dysfunction of hippocampus.
Collapse
Affiliation(s)
| | | | | | | | - Hua Ai
- Institution of Sports Medicine, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
27
|
Murtishaw AS, Heaney CF, Bolton MM, Sabbagh JJ, Langhardt MA, Kinney JW. Effect of acute lipopolysaccharide-induced inflammation in intracerebroventricular-streptozotocin injected rats. Neuropharmacology 2016; 101:110-22. [DOI: 10.1016/j.neuropharm.2015.08.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 08/03/2015] [Accepted: 08/26/2015] [Indexed: 12/25/2022]
|
28
|
Tuttolomondo A, Pecoraro R, Arnao V, Maugeri R, Iacopino DG, Pinto A. Developing drug strategies for the neuroprotective treatment of acute ischemic stroke. Expert Rev Neurother 2015; 15:1271-84. [PMID: 26469760 DOI: 10.1586/14737175.2015.1101345] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Developing new treatment strategies for acute ischemic stroke in the last twenty years has offered some important successes, but also several failures. Most trials of neuroprotective therapies have been uniformly negative to date. Recent research has reported how excitatory amino acids act as the major excitatory neurotransmitters in the cerebral cortex and hippocampus. Furthermore, other therapeutic targets such as free radical scavenger strategies and the anti-inflammatory neuroprotective strategy have been evaluated with conflicting data in animal models and human subjects with acute ischemic stroke. Whereas promising combinations of neuroprotection and neurorecovery, such as citicoline, albumin and cerebrolysin have been tested with findings worthy of further evaluation in larger randomized clinical trials. Understanding the complexities of the ischemic cascade is essential to developing pharmacological targets for acute ischemic stroke in neuroprotective or flow restoration therapeutic strategies.
Collapse
Affiliation(s)
- Antonino Tuttolomondo
- a Internal Medicine and Cardio-Angiology Ward, Department of Biomedicine and Internal Medicine, Di.Bi. M.I.S , University of Palermo , Palermo , Italy
| | - Rosaria Pecoraro
- a Internal Medicine and Cardio-Angiology Ward, Department of Biomedicine and Internal Medicine, Di.Bi. M.I.S , University of Palermo , Palermo , Italy.,b Emergency Care Unit , Fondazione Istituto S. Raffaele/Giglio of Cefalù , Cefalù , Italy
| | - Valentina Arnao
- c Neurology Ward, Department of Experimental Biomedicine and Clinical Neuroscience , University of Palermo , Palermo , Italy
| | - Rosario Maugeri
- d Neurosurgery Ward, Department of Experimental Biomedicine and Clinical Neuroscience , University of Palermo , Palermo , Italy
| | - Domenico Gerardo Iacopino
- d Neurosurgery Ward, Department of Experimental Biomedicine and Clinical Neuroscience , University of Palermo , Palermo , Italy
| | - Antonio Pinto
- a Internal Medicine and Cardio-Angiology Ward, Department of Biomedicine and Internal Medicine, Di.Bi. M.I.S , University of Palermo , Palermo , Italy
| |
Collapse
|
29
|
Liu L, Li CJ, Lu Y, Zong XG, Luo C, Sun J, Guo LJ. Baclofen mediates neuroprotection on hippocampal CA1 pyramidal cells through the regulation of autophagy under chronic cerebral hypoperfusion. Sci Rep 2015; 5:14474. [PMID: 26412641 PMCID: PMC4585985 DOI: 10.1038/srep14474] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/01/2015] [Indexed: 12/13/2022] Open
Abstract
GABA receptors play an important role in ischemic brain injury. Studies have indicated that autophagy is closely related to neurodegenerative diseases. However, during chronic cerebral hypoperfusion, the changes of autophagy in the hippocampal CA1 area, the correlation between GABA receptors and autophagy, and their influences on hippocampal neuronal apoptosis have not been well established. Here, we found that chronic cerebral hypoperfusion resulted in rat hippocampal atrophy, neuronal apoptosis, enhancement and redistribution of autophagy, down-regulation of Bcl-2/Bax ratio, elevation of cleaved caspase-3 levels, reduction of surface expression of GABAA receptor α1 subunit and an increase in surface and mitochondrial expression of connexin 43 (CX43) and CX36. Chronic administration of GABAB receptors agonist baclofen significantly alleviated neuronal damage. Meanwhile, baclofen could up-regulate the ratio of Bcl-2/Bax and increase the activation of Akt, GSK-3β and ERK which suppressed cytodestructive autophagy. The study also provided evidence that baclofen could attenuate the decrease in surface expression of GABAA receptor α1 subunit, and down-regulate surface and mitochondrial expression of CX43 and CX36, which might enhance protective autophagy. The current findings suggested that, under chronic cerebral hypoperfusion, the effects of GABAB receptors activation on autophagy regulation could reverse neuronal damage.
Collapse
Affiliation(s)
- Li Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Chang-jun Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
- Neurology Department, Huanggang central hospital, Hubei Province, Huanggang, 438000, PR China
| | - Yun Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Xian-gang Zong
- Center for Integrated Protein Science (CIPSM) and Zentrum für Pharmaforschung, Department Pharmazie, Ludwig-Maximilians-Universität München, 80539 Munich, Germany
| | - Chao Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Jun Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Lian-jun Guo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
- Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Hubei Province, Wuhan 430030, China
| |
Collapse
|
30
|
Kurzepa J, Szpetnar M, Hordyjewska A, Wątroba S, Gołąb P, Boguszewska-Czubara A. Thrombolytic treatment decreases glutamate/GABA ratio in serum during acute ischaemic stroke: a pilot study. Neurol Res 2015; 37:934-7. [PMID: 26005019 DOI: 10.1179/1743132815y.0000000049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
There is no information about possible effect of recombinant tissue plasminogen activator (rtPA) therapy on excitotoxic/neuroprotective amino acids during acute phase of ischaemic stroke (IS). Our purpose was to evaluate iv thrombolytic treatment on glutamate (Glu) and gamma-aminobutyric acid (GABA) serum levels during acute IS. Eleven thrombolytic (rtPA group) and 12 non-thrombolytic (non-rtPA group) patients with acute IS were enrolled. The serum samples were obtained at three time points for rtPA group (time point 0: first to fourth hour of stroke; time point 1: immediately after rtPA administration; time point 2: on days 5-7 from stroke onset). The remaining patients had blood collection at two time points: time point 1: 5(th)-10(th) hour of stroke and time point 2: on days 5-7 of stroke. Glutamate and GABA were determined by the automated ion-exchange chromatography using Amino Acids Analyser (AAA 400) by INGOS Corp., Praha, Czech Republic. The statistically significant elevation of GABA serum level was noticed directly after thrombolysis (time point 1) in comparison to the corresponding time point in non-rtPA group [0.016 (0.002-0.032) μM/ml vs 0.001 (0.001-0.004) μM/ml for rtPA vs non-rtPA groups, respectively, median (first to third quartile), P < 0.05]. At the same time point, the Glu/GABA ratio was significantly decreased in rtPA group (P < 0.05) suggesting the decrease of excitotoxicity biomarkers in the blood after thrombolysis. Considering the beneficial effect of GABA receptor agonists, the elevation of GABA by rtPA should bring an additional positive features of thrombolytic treatment.
Collapse
|
31
|
Benke D, Balakrishnan K, Zemoura K. Regulation of Cell Surface GABAB Receptors. DIVERSITY AND FUNCTIONS OF GABA RECEPTORS: A TRIBUTE TO HANNS MÖHLER, PART B 2015; 73:41-70. [DOI: 10.1016/bs.apha.2014.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
32
|
Laletin V, Bykov Y. General anesthetics as a factor of effective neuroprotection in ischemic stroke models. ACTA ACUST UNITED AC 2015; 61:440-8. [DOI: 10.18097/pbmc20156104440] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Stroke is the second leading cause of death in the world. Unfortunately, only a few drugs have been proved in clinical trials. Drug development of the last decade has been focused substantially on a promising and heterogeneous group of neuroprotective drugs. Hundreds of compounds were suggested as new putative neuroprotectors, which effectiveness was confirmed in preclinical trials only. At the present time discrepancy between results of preclinical studies and clinical trials requires careful analysis. One of the least evaluated and probably the most noticeable reasons is general anesthesia - an obligatory component of an overwhelming majority of existing animal stroke models. The aim of the review is to describe known mechanisms of common general anesthetics influence on ionotropic and metabotropic plasma membrane receptors, and key signal pathways involved in neuronal hypoxic-ischemic injury and survival
Collapse
Affiliation(s)
- V.S. Laletin
- Irkutsk State Medical University, Irkutsk, Russia
| | - Y.N. Bykov
- Irkutsk State Medical University, Irkutsk, Russia
| |
Collapse
|
33
|
Chernysheva GA, Smol'yakova VI, Osipenko AN, Plotnikov MB. Evaluation of survival and neurological deficit in rats in the new model of global transient cerebral ischemia. Bull Exp Biol Med 2014; 158:197-9. [PMID: 25430646 DOI: 10.1007/s10517-014-2721-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Indexed: 11/24/2022]
Abstract
We propose a modification to rat model of transient global cerebral ischemia with four-vessel occlusion avoiding pneumothorax and minimizing the consequences of surgery. Survival and neurological deficit in rats in this model was studied over 5 days.
Collapse
Affiliation(s)
- G A Chernysheva
- Laboratory of Pharmacology of Blood Circulation, E. D. Goldberg Research Institute of Pharmacology, Siberian Division of the Russian Academy of Medical Sciences, Tomsk, Russia
| | | | | | | |
Collapse
|
34
|
Hodor A, Palchykova S, Baracchi F, Noain D, Bassetti CL. Baclofen facilitates sleep, neuroplasticity, and recovery after stroke in rats. Ann Clin Transl Neurol 2014; 1:765-77. [PMID: 25493268 PMCID: PMC4241804 DOI: 10.1002/acn3.115] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/22/2014] [Accepted: 08/15/2014] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE Sleep disruption in the acute phase after stroke has detrimental effects on recovery in both humans and animals. Conversely, the effect of sleep promotion remains unclear. Baclofen (Bac) is a known non-rapid eye movement (NREM) sleep-promoting drug in both humans and animals. The aim of this study was to investigate the effect of Bac on stroke recovery in a rat model of focal cerebral ischemia (isch). METHODS Rats, assigned to three experimental groups (Bac/isch, saline/isch, or Bac/sham), were injected twice daily for 10 consecutive days with Bac or saline, starting 24 h after induction of stroke. The sleep-wake cycle was assessed by EEG recordings and functional motor recovery by single pellet reaching test (SPR). In order to identify potential neuroplasticity mechanisms, axonal sprouting and neurogenesis were evaluated. Brain damage was assessed by Nissl staining. RESULTS Repeated Bac treatment after ischemia affected sleep, motor function, and neuroplasticity, but not the size of brain damage. NREM sleep amount was increased significantly during the dark phase in Bac/isch compared to the saline/isch group. SPR performance dropped to 0 immediately after stroke and was recovered slowly thereafter in both ischemic groups. However, Bac-treated ischemic rats performed significantly better than saline-treated animals. Axonal sprouting in the ipsilesional motor cortex and striatum, and neurogenesis in the peri-infarct region were significantly increased in Bac/isch group. CONCLUSION Delayed repeated Bac treatment after stroke increased NREM sleep and promoted both neuroplasticity and functional outcome. These data support the hypothesis of the role of sleep as a modulator of poststroke recovery.
Collapse
Affiliation(s)
- Aleksandra Hodor
- Center for Experimental Neurology (ZEN), Department of Neurology, Inselspital, Bern University Hospital 3010, Bern, Switzerland
| | - Svitlana Palchykova
- Center for Experimental Neurology (ZEN), Department of Neurology, Inselspital, Bern University Hospital 3010, Bern, Switzerland
| | - Francesca Baracchi
- Center for Experimental Neurology (ZEN), Department of Neurology, Inselspital, Bern University Hospital 3010, Bern, Switzerland
| | - Daniela Noain
- Department of Neurology, University Hospital Zürich 8091, Zürich, Switzerland
| | - Claudio L Bassetti
- Center for Experimental Neurology (ZEN), Department of Neurology, Inselspital, Bern University Hospital 3010, Bern, Switzerland
| |
Collapse
|
35
|
Xerri C, Zennou-Azogui Y. Early and moderate sensory stimulation exerts a protective effect on perilesion representations of somatosensory cortex after focal ischemic damage. PLoS One 2014; 9:e99767. [PMID: 24914807 PMCID: PMC4051766 DOI: 10.1371/journal.pone.0099767] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/19/2014] [Indexed: 02/05/2023] Open
Abstract
Previous studies have shown that intensive training within an early critical time window after focal cortical ischemia increases the area of damaged tissue and is detrimental to behavioral recovery. We postulated that moderate stimulation initiated soon after the lesion could have protective effects on peri-infarct cortical somatotopic representations. Therefore, we have assessed the effects of mild cutaneous stimulation delivered in an attention-demanding behavioral context on the functional organization of the perilesion somatosensory cortex using high-density electrophysiological mapping. We compared the effects of 6-day training initiated on the 3rd day postlesion (early training; ET) to those of same-duration training started on the 8th day (delayed training; DT). Our findings confirm previous work showing that the absence of training aggravates representational loss in the perilesion zone. In addition, ET was found to be sufficient to limit expansion of the ischemic lesion and reduce tissue loss, and substantially maintain the neuronal responsiveness to tactile stimulation, thereby preserving somatotopic map arrangement in the peri-infarct cortical territories. By contrast, DT did not prevent tissue loss and only partially reinstated lost representations in a use-dependent manner within the spared peri-infarct cortical area. This study differentiates the effects of early versus delayed training on perilesion tissue and cortical map reorganization, and underscores the neuroprotective influence of mild rehabilitative stimulation on neuronal response properties in the peri-infarct cortex during an early critical period.
Collapse
Affiliation(s)
- Christian Xerri
- Neurosciences Intégratives et Adaptatives, Aix-Marseille Université, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7260, Fédération de Recherches Comportement-Cerveau-Cognition 3512, Marseille, France
- * E-mail:
| | - Yoh'i Zennou-Azogui
- Neurosciences Intégratives et Adaptatives, Aix-Marseille Université, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7260, Fédération de Recherches Comportement-Cerveau-Cognition 3512, Marseille, France
| |
Collapse
|
36
|
Li CJ, Lu Y, Zhou M, Zong XG, Li C, Xu XL, Guo LJ, Lu Q. Activation of GABAB receptors ameliorates cognitive impairment via restoring the balance of HCN1/HCN2 surface expression in the hippocampal CA1 area in rats with chronic cerebral hypoperfusion. Mol Neurobiol 2014; 50:704-20. [PMID: 24838625 DOI: 10.1007/s12035-014-8736-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 04/29/2014] [Indexed: 11/30/2022]
Abstract
Hyperpolarization-activated cyclic-nucleotide-gated cation nonselective (HCN) channels are involved in the pathology of nervous system diseases. HCN channels and γ-aminobutyric acid (GABA) receptors can mutually co-regulate the function of neurons in many brain areas. However, little is known about the co-regulation of HCN channels and GABA receptors in the chronic ischemic rats with possible features of vascular dementia. Protein kinase A (PKA) and TPR containing Rab8b interacting protein (TRIP8b) can modulate GABAB receptors cell surface stability and HCN channel trafficking, respectively, and adaptor-associated kinase 1 (AAK1) inhibits the function of the major TRIP8b-interacting protein adaptor protein 2 (AP2) via phosphorylating the AP2 μ2 subunit. Until now, the role of these regulatory factors in chronic cerebral hypoperfusion is unclear. In the present study, we evaluated whether and how HCN channels and GABAB receptors were pathologically altered and investigated neuroprotective effects of GABAB receptors activation and cross-talk networks between GABAB receptors and HCN channels in the hippocampal CA1 area in chronic cerebral hypoperfusion rat model. We found that cerebral hypoperfusion for 5 weeks by permanent occlusion of bilateral common carotid arteries (two-vessel occlusion, 2VO) induced marked spatial and nonspatial learning and memory deficits, significant neuronal loss and decrease in dendritic spine density, impairment of long-term potentiation (LTP) at the Schaffer collateral-CA1 synapses, and reduction of surface expression of GABAB R1, GABAB R2, and HCN1, but increase in HCN2 surface expression. Meanwhile, the protein expression of TRIP8b (1a-4), TRIP8b (1b-2), and AAK1 was significantly decreased. Baclofen, a GABAB receptor agonist, markedly improved the memory impairment and alleviated neuronal damage. Besides, baclofen attenuated the decrease of surface expression of GABAB R1, GABAB R2, and HCN1, but downregulated HCN2 surface expression. Furthermore, baclofen could restore expression of AAK1 protein and significantly increase p-PKA, TRIP8b (1a-4), TRIP8b (1b-2), and p-AP2 μ2 expression. Those findings suggested that, under chronic cerebral hypoperfusion, activation of PKA could attenuate baclofen-induced decrease in surface expression of GABAB R1 and GABAB R2, and activation of GABAB receptors not only increased the expression of TRIP8b (1a-4) and TRIP8b (1b-2) but also regulated the function of TRIP8b via AAK1 and p-AP2 μ2, which restored the balance of HCN1/HCN2 surface expression in rat hippocampal CA1 area, and thus ameliorated cognitive impairment.
Collapse
Affiliation(s)
- Chang-jun Li
- Department of Pharmacology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Di Vito A, Mele M, Piscioneri A, Morelli S, Bartolo LD, Barni T, Facciolo RM, Canonaco M. Overstimulation of glutamate signals leads to hippocampal transcriptional plasticity in hamsters. Cell Mol Neurobiol 2014; 34:501-9. [PMID: 24532154 PMCID: PMC11488877 DOI: 10.1007/s10571-014-0034-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/28/2014] [Indexed: 01/07/2023]
Abstract
It's known that neurons in mammalian hibernators are more tolerant to hypoxia than those in non-hibernating species and as a consequence animals are capable of awakening from the arousal state without exhibiting cerebral damages. In addition, evidences have suggested that euthermic hamster neurons display protective adaptations against hypoxia, while those of rats are not capable, even though molecular mechanisms involved in similar neuroprotective strategies have not been yet fully studied. In the present work, overstimulation of glutamatergic receptors NMDA recognized as one of the major death-promoting element in hypoxia, accounted for altered network complexity consistent with a moderate reduction of hippocampal neuronal survival (p < 0.05) in hamsters. These alterations appeared to be featured concomitantly with altered glutamatergic signaling as indicated by significant down-regulation (p < 0.01) of NMDAergic (NR2A) and AMPAergic (GluR1, R2) receptor subtypes together with the metabotropic mGluR5 subtype. Diminished mRNA levels were also reported for NMDA receptor binding factors and namely PSD95 plus DREAM, which exert positive and negative regulatory properties, respectively, on receptor trafficking events. Conversely, involvement of glutamatergic signaling systems on neuronal excitotoxicity was strengthened by the co-activation of GABAAR-mediated effects as indicated by toxic morphological effects being notably reduced along with up-regulated GluR1, GluR2, mGluR5, DREAM, and Homer1c scaffold proteins when muscimol was added. Overall, these results point to a neuroprotective role of the GABAergic system against excitotoxicity episodes via DREAM-dependent inhibition of NMDA receptor and activation of AMPA receptor plus mGluR5, respectively, thus proposing them as novel therapeutic targets against cerebral ischemic damages in humans.
Collapse
Affiliation(s)
- Anna Di Vito
- Molecular Oncology Laboratory, Experimental and Clinical Medicine Department, University Magna Graecia of Catanzaro, 88100, Catanzaro, Italy,
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Maier PJ, Zemoura K, Acuña MA, Yévenes GE, Zeilhofer HU, Benke D. Ischemia-like oxygen and glucose deprivation mediates down-regulation of cell surface γ-aminobutyric acidB receptors via the endoplasmic reticulum (ER) stress-induced transcription factor CCAAT/enhancer-binding protein (C/EBP)-homologous protein (CHOP). J Biol Chem 2014; 289:12896-907. [PMID: 24668805 DOI: 10.1074/jbc.m114.550517] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cerebral ischemia frequently leads to long-term disability and death. Excitotoxicity is believed to be the main cause for ischemia-induced neuronal death. Although a role of glutamate receptors in this process has been firmly established, the contribution of metabotropic GABAB receptors, which control excitatory neurotransmission, is less clear. A prominent characteristic of ischemic insults is endoplasmic reticulum (ER) stress associated with the up-regulation of the transcription factor CCAAT/enhancer-binding protein-homologous protein (CHOP). After inducing ER stress in cultured cortical neurons by sustained Ca(2+) release from intracellular stores or by a brief episode of oxygen and glucose deprivation (in vitro model of cerebral ischemia), we observed an increased expression of CHOP accompanied by a strong reduction of cell surface GABAB receptors. Our results indicate that down-regulation of cell surface GABAB receptors is caused by the interaction of the receptors with CHOP in the ER. Binding of CHOP prevented heterodimerization of the receptor subunits GABAB1 and GABAB2 and subsequent forward trafficking of the receptors to the cell surface. The reduced level of cell surface receptors diminished GABAB receptor signaling and, thus, neuronal inhibition. These findings indicate that ischemia-mediated up-regulation of CHOP down-regulates cell surface GABAB receptors by preventing their trafficking from the ER to the plasma membrane. This mechanism leads to diminished neuronal inhibition and may contribute to excitotoxicity in cerebral ischemia.
Collapse
Affiliation(s)
- Patrick J Maier
- From the Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
39
|
Xu C, Zhang W, Rondard P, Pin JP, Liu J. Complex GABAB receptor complexes: how to generate multiple functionally distinct units from a single receptor. Front Pharmacol 2014; 5:12. [PMID: 24575041 PMCID: PMC3920572 DOI: 10.3389/fphar.2014.00012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/22/2014] [Indexed: 01/05/2023] Open
Abstract
The main inhibitory neurotransmitter, GABA, acts on both ligand-gated and G protein-coupled receptors, the GABAA/C and GABAB receptors, respectively. The later play important roles in modulating many synapses, both at the pre- and post-synaptic levels, and are then still considered as interesting targets to treat a number of brain diseases, including addiction. For many years, several subtypes of GABAB receptors were expected, but cloning revealed only two genes that work in concert to generate a single type of GABAB receptor composed of two subunits. Here we will show that the signaling complexity of this unit receptor type can be largely increased through various ways, including receptor stoichiometry, subunit isoforms, cell-surface expression and localization, crosstalk with other receptors, or interacting proteins. These recent data revealed how complexity of a receptor unit can be increased, observation that certainly are not unique to the GABAB receptor.
Collapse
Affiliation(s)
- Chanjuan Xu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan, China
| | - Wenhua Zhang
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan, China
| | - Philippe Rondard
- Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U661, Universités de Montpellier I & II Montpellier, France
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U661, Universités de Montpellier I & II Montpellier, France
| | - Jianfeng Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology Wuhan, China
| |
Collapse
|
40
|
Huang CY, Kuo WW, Wang HF, Lin CJ, Lin YM, Chen JL, Kuo CH, Chen PK, Lin JY. GABA tea ameliorates cerebral cortex apoptosis and autophagy in streptozotocin-induced diabetic rats. J Funct Foods 2014. [DOI: 10.1016/j.jff.2013.11.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
41
|
Postresuscitation Treatment With Argon Improves Early Neurological Recovery in a Porcine Model of Cardiac Arrest. Shock 2014; 41:72-8. [DOI: 10.1097/shk.0000000000000049] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Benke D. GABAB receptor trafficking and interacting proteins: Targets for the development of highly specific therapeutic strategies to treat neurological disorders? Biochem Pharmacol 2013; 86:1525-30. [DOI: 10.1016/j.bcp.2013.09.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 09/20/2013] [Accepted: 09/20/2013] [Indexed: 10/26/2022]
|
43
|
Caceres LG, Cid MP, Uran SL, Zorrilla Zubilete MA, Salvatierra NA, Guelman LR. Pharmacological alterations that could underlie radiation-induced changes in associative memory and anxiety. Pharmacol Biochem Behav 2013; 111:37-43. [PMID: 23958578 DOI: 10.1016/j.pbb.2013.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/02/2013] [Accepted: 08/08/2013] [Indexed: 11/30/2022]
Abstract
It is widely known that ionizing radiation is a physical agent broadly used to kill tumor cells during human cancer therapy. Unfortunately, adjacent normal tissues can concurrently undergo undesirable cell injury. Previous data of our laboratory demonstrated that exposure of developing rats to ionizing radiations induced a variety of behavioral differences respect to controls, including changes in associative memory and in anxiety state. However, there is a lack of data concerning modifications in different related pharmacological intermediaries. Therefore, the aim of the present study was to investigate whether the behavioral differences observed in young animals irradiated at birth might be underlain by early changes in PKCß1 levels which, in turn, could lead to changes in hippocampal GABAergic neurotransmission. Male Wistar rats were irradiated with 5Gy of X rays between 24 and 48 h after birth. Different pharmacological markers related to the affected behavioral tasks were assessed in control and irradiated hippocampus at 15 and 30 days, namely GABAA receptor, GAD65-67, ROS and PKCß1. Results showed that all measured parameters were increased in the hippocampus of 30-days-old irradiated animals. In contrast, in the hippocampus of 15-days-old irradiated animals only the levels of PKCß1 were decreased. These data suggest that PKCß1 might constitute a primary target for neonatal radiation damage on the hippocampus. Therefore, it could be hypothesized that an initial decrease in the levels of this protein can trigger a subsequent compensatory increase that, in turn, could be responsible for the plethora of biochemical changes that might underlie the previously observed behavioral alterations.
Collapse
Affiliation(s)
- L G Caceres
- 1ª Cátedra de Farmacología, Facultad de Medicina, UBA-CEFyBO-CONICET, Paraguay 2155, piso 15, (1121) Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
44
|
Paula-Lima AC, Brito-Moreira J, Ferreira ST. Deregulation of excitatory neurotransmission underlying synapse failure in Alzheimer's disease. J Neurochem 2013; 126:191-202. [PMID: 23668663 DOI: 10.1111/jnc.12304] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 05/09/2013] [Accepted: 05/10/2013] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly. Memory loss in AD is increasingly attributed to soluble oligomers of the amyloid-β peptide (AβOs), toxins that accumulate in AD brains and target particular synapses. Glutamate receptors appear to be centrally involved in synaptic targeting by AβOs. Once bound to neurons, AβOs dysregulate the activity and reduce the surface expression of both N-methyl-D-aspartate (NMDA) and 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl)propanoic acid (AMPA) types of glutamate receptors, impairing signaling pathways involved in synaptic plasticity. In the extracellular milieu, AβOs promote accumulation of the excitatory amino acids, glutamate and D-serine. This leads to overactivation of glutamate receptors, triggering abnormal calcium signals with noxious impacts on neurons. Here, we review key findings linking AβOs to deregulated glutamate neurotransmission and implicating this as a primary mechanism of synapse failure in AD. We also discuss strategies to counteract the impact of AβOs on excitatory neurotransmission. In particular, we review evidence showing that inducing neuronal hyperpolarization via activation of inhibitory GABA(A) receptors prevents AβO-induced excitotoxicity, suggesting that this could comprise a possible therapeutic approach in AD.
Collapse
Affiliation(s)
- Andrea C Paula-Lima
- Department of Basic Sciences, Faculty of Dentistry, University of Chile, Santiago, Chile.
| | | | | |
Collapse
|
45
|
Grönbladh A, Johansson J, Nyberg F, Hallberg M. Recombinant human growth hormone affects the density and functionality of GABAB receptors in the male rat brain. Neuroendocrinology 2013; 97:203-11. [PMID: 22710737 DOI: 10.1159/000339821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Accepted: 05/27/2012] [Indexed: 11/19/2022]
Abstract
The beneficial effects of growth hormone (GH) on memory and learning have previously been confirmed in both humans and in animal models. An important role of GABAB receptors for multiple forms of learning and memory has also been reported. In this study, we examined the effect of GH on the density and functionality of the metabotropic GABAB receptors in the rat brain. Male Sprague-Dawley rats (n = 24) divided into 3 groups were injected twice daily with recombinant human GH (0.07 or 0.7 IU/kg) for 7 days. The effects of the hormone were determined by quantitative autoradiography and by GABAB stimulated [(35)S]-GTPγS binding using the selective GABAB receptor agonist baclofen. The results demonstrate moderate but significant alterations in both receptor density and functionality in a number of brain regions. For example, a dose-dependent upregulation of GABAB receptors was found in the cingulate cortex, primary motor cortex and caudate putamen, whereas attenuation in the receptor density was encountered in, for example, the medial geniculate nucleus. Although the GH-induced effects on the GABAB receptor in brain areas associated with cognition were fairly pronounced, they were significant and we propose that the physiological responses observed after GH administration at least partly can be mediated through a mechanism involving GABAB receptors.
Collapse
Affiliation(s)
- Alfhild Grönbladh
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | | |
Collapse
|
46
|
Montori S, DosAnjos S, Poole A, Regueiro-Purriños MM, Llorente IL, Darlison MG, Fernández-López A, Martínez-Villayandre B. Differential effect of transient global ischaemia on the levels of γ-aminobutyric acid type A (GABAA) receptor subunit mRNAs in young and older rats. Neuropathol Appl Neurobiol 2012; 38:710-22. [DOI: 10.1111/j.1365-2990.2012.01254.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
47
|
Zhu J, Shao CY, Yang W, Zhang XM, Wu ZY, Zhou L, Wang XX, Li YH, Xia J, Luo JH, Shen Y. Chronic zinc exposure decreases the surface expression of NR2A-containing NMDA receptors in cultured hippocampal neurons. PLoS One 2012; 7:e46012. [PMID: 23049922 PMCID: PMC3457937 DOI: 10.1371/journal.pone.0046012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 08/23/2012] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Zinc distributes widely in the central nervous system, especially in the hippocampus, amygdala and cortex. The dynamic balance of zinc is critical for neuronal functions. Zinc modulates the activity of N-methyl-D-aspartate receptors (NMDARs) through the direct inhibition and various intracellular signaling pathways. Abnormal NMDAR activities have been implicated in the aetiology of many brain diseases. Sustained zinc accumulation in the extracellular fluid is known to link to pathological conditions. However, the mechanism linking this chronic zinc exposure and NMDAR dysfunction is poorly understood. METHODOLOGY/PRINCIPAL FINDINGS We reported that chronic zinc exposure reduced the numbers of NR1 and NR2A clusters in cultured hippocampal pyramidal neurons. Whole-cell and synaptic NR2A-mediated currents also decreased. By contrast, zinc did not affect NR2B, suggesting that chronic zinc exposure specifically influences NR2A-containg NMDARs. Surface biotinylation indicated that zinc exposure attenuated the membrane expression of NR1 and NR2A, which might arise from to the dissociation of the NR2A-PSD-95-Src complex. CONCLUSIONS Chronic zinc exposure perturbs the interaction of NR2A to PSD-95 and causes the disorder of NMDARs in hippocampal neurons, suggesting a novel action of zinc distinct from its acute effects on NMDAR activity.
Collapse
Affiliation(s)
- Jia Zhu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Chong-Yu Shao
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Wei Yang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Xiao-Min Zhang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Zhen-Yong Wu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Liang Zhou
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Xin-Xin Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Yun-Hong Li
- Department of Neurobiology, Center of Scientific Technology, Cranial Cerebral Disease Lab, Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Jun Xia
- Department of Biochemistry, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, People’s Republic of China
| | - Jian-Hong Luo
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Ying Shen
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| |
Collapse
|
48
|
Wei XW, Yan H, Xu B, Wu YP, Li C, Zhang GY. Neuroprotection of co-activation of GABA receptors by preventing caspase-3 denitrosylation in KA-induced seizures. Brain Res Bull 2012; 88:617-23. [PMID: 22613773 DOI: 10.1016/j.brainresbull.2012.05.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 05/07/2012] [Accepted: 05/09/2012] [Indexed: 12/19/2022]
Abstract
Previous studies have demonstrated that kainic acid (KA)-induced seizures can cause the enhancement of excitation and lead to neuronal death in rat hippocampus. Co-activation of the inhibitory GABA receptors can attenuate the excitatory JNK3 apoptotic signaling pathway via inhibiting the increased assembly of the GluR6-PSD-95-MLK3 signaling module induced by KA in epileptic rat hippocampal CA1 and CA3 regions. Caspase-3 is a cysteine protease located in both the cytoplasm and mitochondrial intermembrane space that is a central effector of many apoptotic pathways. We designed experiments to elucidate the underlying molecular mechanisms of procaspase-3 activation and neuroprotection of co-activation of GABA receptors against neuronal death induced by KA. In this study, we show that co-activation of GABA receptors can attenuate the Fas/FasL apoptotic signaling pathway and inhibit the increased of thioredoxin reductase activity induced by KA, subsequently inhibit the activation of procaspase-3 by diminishing the denitrosylation of its active-site thiol and decreasing the cleavage of the caspase-3 zymogen to its active subunits. These results indicate that co-activation of GABA receptors results in neuroprotection by preventing caspase-3 denitrosylation in KA-induced seizure of rats.
Collapse
Affiliation(s)
- Xue-Wen Wei
- Jiangsu Key Laboratory of Brain Disease Bioinformation and Research Center of Biochemistry and Molecular Biochemistry, Xuzhou Medical College, Xuzhou 221002, China
| | | | | | | | | | | |
Collapse
|
49
|
Baclofen influences acquisition and MMP-2, MMP-9 levels in the hippocampus of rats after hypoxia. Pharmacol Rep 2012; 64:536-45. [DOI: 10.1016/s1734-1140(12)70849-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 02/02/2012] [Indexed: 11/18/2022]
|
50
|
Tang LJ, Li C, Hu SQ, Wu YP, Zong YY, Sun CC, Zhang F, Zhang GY. S-nitrosylation of c-Src via NMDAR-nNOS module promotes c-Src activation and NR2A phosphorylation in cerebral ischemia/reperfusion. Mol Cell Biochem 2012; 365:363-77. [PMID: 22422045 DOI: 10.1007/s11010-012-1280-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 03/02/2012] [Indexed: 10/28/2022]
Abstract
Previous studies suggested that activated c-Src promote the tyrosine phosphorylation of NMDA receptor subunit NR2A, and thus aggravate the injury induced by transient cerebral ischemia/reperfusion (I/R) in rat hippocampus CA1 region. In this study, we examined the effect of nitric oxide (NO) on the activation of c-Src and the tyrosine phosphorylation of NMDA receptor NR2A subunit. The results show that S-nitrosylation and the phosphorylation of c-Src were induced after cerebral I/R in rats, and administration of nNOS inhibitor 7-NI, nNOS antisense oligonucleotides and exogenous NO donor sodium nitroprusside diminished the increased S-nitrosylation and phosphorylation of c-Src during cerebral I/R. The cysteine residues of c-Src modified by S-nitrosylation are Cys489, Cys498, and Cys500. On the other hand, NMDAR antagonist MK-801 could attenuate the S-nitrosylation and activation of c-Src. Taken together, the S-nitrosylation of c-Src is provoked by NO derived from endogenous nNOS, which is activated by Ca(2+) influx from NMDA receptors, and promotes the auto-phosphorylation at tyrosines and further phosphorylates NR2A. The molecular mechanism we outlined here is a novel postsynaptic NMDAR-nNOS/c-Src-mediated signaling amplification, the 'NMDAR-nNOS → NO → SNO-c-Src → p-c-Src → NMDAR-nNOS' cycle, which presents the possibility as a potential therapeutic target for stroke treatment.
Collapse
Affiliation(s)
- Li-Juan Tang
- Research Center of Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou 221002, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|