1
|
Zepeda-Morales K, Bravo D, Aránguiz-Barrera J, Ampuero E, Renard GM, Pelissier T, Hernández A, Retamal JS, Constandil L. N-methyl-D-aspartate receptor activation is downstream coupled to pannexin 1 opening by Src kinase in dorsal horn neurons: an essential link for mechanical hyperalgesia in nerve-injured rats. Pain 2025; 166:1369-1381. [PMID: 39630026 PMCID: PMC12067615 DOI: 10.1097/j.pain.0000000000003476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/24/2024] [Accepted: 10/07/2024] [Indexed: 05/14/2025]
Abstract
ABSTRACT A well-recognized molecular entity involved in pain-related neuroplasticity is the N-methyl-D-aspartate receptor (NMDAR), which is crucial for developing chronic pain. Likewise, the pannexin 1 (Panx1) channel has been described as necessary for initiating and maintaining neuropathic pain, driving nociceptive signals dependent on spinal NMDAR through several possible mechanisms. Through behavioral, pharmacological, and molecular approaches, our study in male rats has revealed several key findings: (1) neurons located in spinal cord laminae I and II express functional Panx1 channels in both neuropathic and sham rats. These channels can open (indicated by YOPRO-1 uptake) through the stimulation of NMDARs with intrathecal NMDA; (2) intrathecal NMDA leads to increased expression of pSrc and pPanx1 in dorsal horn neurons. This elevation exacerbates existing mechanical hyperalgesia in nerve-injured rats; (3) inhibition of Src with intrathecal PP2 or blockade of Panx1 with intrathecal 10 Panx effectively mitigates NMDA-induced effects and reduces the spontaneous mechanical hyperalgesia of nerve-injured rats. Notably, while 10 Panx successfully alleviates hyperalgesia, it does not alter pSrc expression; and (4) NMDA-stimulated YOPRO-1 uptake in neurons of laminae I-II of spinal cord slices were prevented by the NMDAR antagonist D-AP5, the Src inhibitor PP2 (but not PP3), as well as with the 10 Panx and carbenoxolone. Therefore, NMDAR activation in dorsal horn neurons triggers an NMDAR-Src-Panx1 signaling pathway, where Panx1 acts as an enhancing effector in neuropathic pain. This implies that disrupting the NMDAR-Panx1 communication (eg, through Src inhibitors and/or Panx1 blockers) may offer a valuable strategy for managing some forms of chronic pain.
Collapse
Affiliation(s)
- Katherine Zepeda-Morales
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - David Bravo
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Jonathan Aránguiz-Barrera
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Estibaliz Ampuero
- Laboratory of Neurobiology of Behavior, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Georgina M. Renard
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Teresa Pelissier
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Alejandro Hernández
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Jeffri S. Retamal
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Luis Constandil
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| |
Collapse
|
2
|
Chen R, Yi Y, Xiao W, Zhong B, Zhang L, Zeng Y. Urinary protein biomarkers based on LC-MS/MS analysis to discriminate vascular dementia from Alzheimer's disease in Han Chinese population. Front Aging Neurosci 2023; 15:1070854. [PMID: 36761180 PMCID: PMC9905227 DOI: 10.3389/fnagi.2023.1070854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023] Open
Abstract
Objective This study aimed to identify the potential urine biomarkers of vascular dementia (VD) and unravel the disease-associated mechanisms by applying Liquid chromatography tandem-mass spectrometry (LC-MS/MS). Methods LC-MS/MS proteomic analysis was applied to urine samples from 3 groups, including 14 patients with VD, 9 patients with AD, and 21 normal controls (NC). By searching the MS data by Proteome Discoverer software, analyzing the protein abundances qualitatively and quantitatively, comparing between groups, combining bioinformatics analysis using Gene Ontology (GO) and pathway crosstalk analysis using Kyoto Encyclopedia of Genes and Genomes (KEGG), and literature searching, the differentially expressed proteins (DEPs) of VD can be comprehensively determined at last and were further quantified by receiver operating characteristic (ROC) curve methods. Results The proteomic findings showed quantitative changes in patients with VD compared to patients with NC and AD groups; among 4,699 identified urine proteins, 939 and 1,147 proteins displayed quantitative changes unique to VD vs. NC and AD, respectively, including 484 overlapped common DEPs. Then, 10 unique proteins named in KEGG database (including PLOD3, SDCBP, SRC, GPRC5B, TSG101/STP22/VPS23, THY1/CD90, PLCD, CDH16, NARS/asnS, AGRN) were confirmed by a ROC curve method. Conclusion Our results suggested that urine proteins enable detection of VD from AD and VC, which may provide an opportunity for intervention.
Collapse
Affiliation(s)
- Ruijuan Chen
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China,Department of Emergency, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan, China
| | - Yuanjing Yi
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenbiao Xiao
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bowen Zhong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Le Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Zeng
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China,*Correspondence: Yi Zeng,
| |
Collapse
|
3
|
de Pins B, Mendes T, Giralt A, Girault JA. The Non-receptor Tyrosine Kinase Pyk2 in Brain Function and Neurological and Psychiatric Diseases. Front Synaptic Neurosci 2021; 13:749001. [PMID: 34690733 PMCID: PMC8527176 DOI: 10.3389/fnsyn.2021.749001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/14/2021] [Indexed: 12/28/2022] Open
Abstract
Pyk2 is a non-receptor tyrosine kinase highly enriched in forebrain neurons. Pyk2 is closely related to focal adhesion kinase (FAK), which plays an important role in sensing cell contacts with extracellular matrix and other extracellular signals controlling adhesion and survival. Pyk2 shares some of FAK’s characteristics including recruitment of Src-family kinases after autophosphorylation, scaffolding by interacting with multiple partners, and activation of downstream signaling pathways. Pyk2, however, has the unique property to respond to increases in intracellular free Ca2+, which triggers its autophosphorylation following stimulation of various receptors including glutamate NMDA receptors. Pyk2 is dephosphorylated by the striatal-enriched phosphatase (STEP) that is highly expressed in the same neuronal populations. Pyk2 localization in neurons is dynamic, and altered following stimulation, with post-synaptic and nuclear enrichment. As a signaling protein Pyk2 is involved in multiple pathways resulting in sometimes opposing functions depending on experimental models. Thus Pyk2 has a dual role on neurites and dendritic spines. With Src family kinases Pyk2 participates in postsynaptic regulations including of NMDA receptors and is necessary for specific types of synaptic plasticity and spatial memory tasks. The diverse functions of Pyk2 are also illustrated by its role in pathology. Pyk2 is activated following epileptic seizures or ischemia-reperfusion and may contribute to the consequences of these insults whereas Pyk2 deficit may contribute to the hippocampal phenotype of Huntington’s disease. Pyk2 gene, PTK2B, is associated with the risk for late-onset Alzheimer’s disease. Studies of underlying mechanisms indicate a complex contribution with involvement in amyloid toxicity and tauopathy, combined with possible functional deficits in neurons and contribution in microglia. A role of Pyk2 has also been proposed in stress-induced depression and cocaine addiction. Pyk2 is also important for the mobility of astrocytes and glioblastoma cells. The implication of Pyk2 in various pathological conditions supports its potential interest for therapeutic interventions. This is possible through molecules inhibiting its activity or increasing it through inhibition of STEP or other means, depending on a precise evaluation of the balance between positive and negative consequences of Pyk2 actions.
Collapse
Affiliation(s)
- Benoit de Pins
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Tiago Mendes
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Jean-Antoine Girault
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| |
Collapse
|
4
|
Chen H, Chen X, Luo Y, Shen J. Potential molecular targets of peroxynitrite in mediating blood–brain barrier damage and haemorrhagic transformation in acute ischaemic stroke with delayed tissue plasminogen activator treatment. Free Radic Res 2018; 52:1220-1239. [PMID: 30468092 DOI: 10.1080/10715762.2018.1521519] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hansen Chen
- School of Chinese Medicine, the University of Hong Kong, PR China
- Shenzhen Institute of Research and Innovation (HKU-SIRI), University of Hong Kong, Hong Kong, PR China
| | - Xi Chen
- Department of Core Facility, the People’s Hospital of Bao-an Shenzhen, Shenzhen, PR China
- The 8th People’s Hospital of Shenzhen, the Affiliated Bao-an Hospital of Southern Medical University, Shenzhen, PR China
| | - Yunhao Luo
- School of Chinese Medicine, the University of Hong Kong, PR China
| | - Jiangang Shen
- School of Chinese Medicine, the University of Hong Kong, PR China
- Shenzhen Institute of Research and Innovation (HKU-SIRI), University of Hong Kong, Hong Kong, PR China
| |
Collapse
|
5
|
Lu F, Shao G, Wang Y, Guan S, Burlingame AL, Liu X, Liang X, Knox R, Ferriero DM, Jiang X. Hypoxia-ischemia modifies postsynaptic GluN2B-containing NMDA receptor complexes in the neonatal mouse brain. Exp Neurol 2017; 299:65-74. [PMID: 28993251 DOI: 10.1016/j.expneurol.2017.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/09/2017] [Accepted: 10/05/2017] [Indexed: 01/08/2023]
Abstract
The N-methyl-d-aspartate-type glutamate receptor (NMDAR)-associated multiprotein complexes are indispensable for synaptic plasticity and cognitive functions. While purification and proteomic analyses of these signaling complexes have been performed in adult rodent and human brain, much less is known about the protein composition of NMDAR complexes in the developing brain and their modifications by neonatal hypoxic-ischemic (HI) brain injury. In this study, the postsynaptic density proteins were prepared from postnatal day 9 naïve, sham-operated and HI-injured mouse cortex. The GluN2B-containing NMDAR complexes were purified by immunoprecipitation with a mouse GluN2B antibody and subjected to mass spectrometry analysis for determination of the GluN2B binding partners. A total of 71 proteins of different functional categories were identified from the naïve animals as native GluN2B-interacting partners in the developing mouse brain. Neonatal HI reshaped the postsynaptic GluN2B interactome by recruiting new proteins, including multiple kinases, into the complexes; and modifying the existing associations within 1h of reperfusion. The early responses of postsynaptic NMDAR complexes and their related signaling networks may contribute to molecular processes leading to cell survival or death, brain damage and/or neurological disorders in term infants with neonatal encephalopathy.
Collapse
Affiliation(s)
- Fuxin Lu
- Department of Pediatrics, University of California San Francisco, CA, USA
| | - Guo Shao
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
| | - Yongqiang Wang
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, CA, USA; Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Shenheng Guan
- Department of Pharmaceutical Chemistry, University of California San Francisco, CA, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California San Francisco, CA, USA
| | - Xuemei Liu
- Central Laboratory, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Liang
- Central Laboratory, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Renatta Knox
- Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA
| | - Donna M Ferriero
- Department of Pediatrics, University of California San Francisco, CA, USA; Department of Neurology, University of California San Francisco, CA, USA
| | - Xiangning Jiang
- Department of Pediatrics, University of California San Francisco, CA, USA.
| |
Collapse
|
6
|
Role of Phosphorylated HDAC4 in Stroke-Induced Angiogenesis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2957538. [PMID: 28127553 PMCID: PMC5239970 DOI: 10.1155/2017/2957538] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/01/2016] [Indexed: 12/30/2022]
Abstract
Acetylation or deacetylation of chromatin proteins and transcription factors is part of a complex signaling system that is involved in the control of neurological disorders. Recent studies have demonstrated that histone deacetylases (HDACs) exert protective effects in attenuating neuronal injury after ischemic insults. Class IIa HDAC4 is highly expressed in the brain, and neuronal activity depends on the nucleocytoplasmic shuttling of HDAC4. However, little is known about HDAC4 and its roles in ischemic stroke. In this study, we report that phosphorylation of HDAC4 was remarkably upregulated after stroke and blockade of HDAC4 phosphorylation with GÖ6976 repressed stroke-induced angiogenesis. Phosphorylation of HDAC4 was also increased in endothelial cells hypoxia model and suppression of HDAC4 phosphorylation inhibited the tube formation and migration of endothelial cells in vitro. Furthermore, in addition to the inhibition of angiogenesis, blockade of HDAC4 phosphorylation suppressed the expression of genes downstream of HIF-VEGF signaling in vitro and in vivo. These data indicate that phosphorylated HDAC4 may serve as an important regulator in stroke-induced angiogenesis. The protective mechanism of phosphorylated HDAC4 is associated with HIF-VEGF signaling, implicating a novel therapeutic target in stroke.
Collapse
|
7
|
Clonidine preconditioning alleviated focal cerebral ischemic insult in rats via up-regulating p-NMDAR1 and down-regulating NMDAR2A / p-NMDAR2B. Eur J Pharmacol 2016; 793:89-94. [PMID: 27806917 DOI: 10.1016/j.ejphar.2016.10.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 10/22/2016] [Accepted: 10/24/2016] [Indexed: 01/17/2023]
Abstract
A brain ischemia rat model was established by middle cerebral artery occlusion (MCAO) for 2h and reperfusion for 4h to investigate the underlying mechanism of the neuroprotection action of clonidine, a classical alpha-2 adrenergic agonist, on cerebral ischemia. Clonidine and yohimbine were intraperitoneally given to the rats each day for a week before ischemia. Neurological deficits evaluations were carried out at 6h after operation. TTC staining method was used to measure the volume of brain infarction. Expression levels of NMDAR1, NMDAR2A, NMDAR2B were assayed by western blotting. Our data demonstrated that clonidine pretreatment significantly improved the neurological deficit scores and reduced the brain infarct volumes of the rats. Furthermore, protein expression level of p-NMDAR2B in cortex was significantly up-regulated whereas that of p-NMDAR1 was decreased when compared with the sham-operated rats. Remarkably, clonidine treatment led to significant down-regulation of p-NMDAR2B and NMDAR2A in addition to enhancement of the expression level of p-NMDAR1 in cortex. This is the first report illustrating the neuroprotective role of clonidine may be mediated through modulation of the expression levels of p-NMDAR2B, NMDAR2A and p-NMDAR1 during cerebral ischemia.
Collapse
|
8
|
Zhao EY, Efendizade A, Cai L, Ding Y. The role of Akt (protein kinase B) and protein kinase C in ischemia-reperfusion injury. Neurol Res 2016; 38:301-8. [PMID: 27092987 DOI: 10.1080/01616412.2015.1133024] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Stroke is a leading cause of long-term disability and death in the United States. Currently, tissue plasminogen activator (tPA), is the only Food and Drug Administration-approved treatment for acute ischemic stroke. However, the use of tPA is restricted to a small subset of acute stroke patients due to its limited 3-h therapeutic time window. Given the limited therapeutic options at present and the multi-factorial progression of ischemic stroke, emphasis has been placed on the discovery and use of combination therapies aimed at various molecular targets contributing to ischemic cell death. Protein kinase C (PKC) and Akt (protein kinase B) are serine/threonine kinases that play a critical role in mediating ischemic-reperfusion injury and cellular growth and survival, respectively. The present review will examine the role of PKC and Akt in the cellular response to ischemic-reperfusion injury.
Collapse
Affiliation(s)
- Ethan Y Zhao
- a Departmentof Neurosurgery , Wayne State University School of Medicine , Detroit , MI 48201 , USA
| | - Aslan Efendizade
- b Michigan State University College of Osteopathic Medicine , East Lansing , MI 48825 , USA
| | - Lipeng Cai
- c Department of Neurology , China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University , Beijing , China
| | - Yuchuan Ding
- a Departmentof Neurosurgery , Wayne State University School of Medicine , Detroit , MI 48201 , USA.,c Department of Neurology , China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University , Beijing , China
| |
Collapse
|
9
|
The Functional and Molecular Properties, Physiological Functions, and Pathophysiological Roles of GluN2A in the Central Nervous System. Mol Neurobiol 2016; 54:1008-1021. [DOI: 10.1007/s12035-016-9715-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/11/2016] [Indexed: 11/25/2022]
|
10
|
Zhang Z, Wang Y, Yan S, Du F, Yan SS. NR2B-dependent cyclophilin D translocation suppresses the recovery of synaptic transmission after oxygen-glucose deprivation. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2225-2234. [PMID: 26232180 DOI: 10.1016/j.bbadis.2015.07.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 07/24/2015] [Accepted: 07/27/2015] [Indexed: 02/08/2023]
Abstract
N-methyl d-aspartate receptor (NMDA) subunit 2B (NR2B)-containing NMDA receptors and mitochondrial protein cyclophilin D (CypD) are well characterized in mediating neuronal death after ischemia, respectively. However, whether and how NR2B and CypD work together in mediating synaptic injury after ischemia remains elusive. Using an ex vivo ischemia model of oxygen-glucose deprivation (OGD) in hippocampal slices, we identified a NR2B-dependent mechanism for CypD translocation onto the mitochondrial inner membrane. CypD depletion (CypD null mice) prevented OGD-induced impairment in synaptic transmission recovery. Overexpression of neuronal CypD mice (CypD+) exacerbated OGD-induced loss of synaptic transmission. Inhibition of CypD-dependent mitochondrial permeability transition pore (mPTP) opening by cyclosporine A (CSA) attenuated ischemia-induced synaptic perturbation in CypD+ and non-transgenic (non-Tg) mice. The treatment of antioxidant EUK134 to suppress mitochondrial oxidative stress rescued CypD-mediated synaptic dysfunction following OGD in CypD+ slices. Furthermore, OGD provoked the interaction of CypD with P53, which was enhanced in slices overexpressing CypD but was diminished in CypD-null slices. Inhibition of p53 using a specific inhibitor of p53 (pifithrin-μ) attenuated the CypD/p53 interaction following OGD, along with a restored synaptic transmission in both non-Tg and CypD+ hippocampal slices. Our results indicate that OGD-induced CypD translocation potentiates CypD/P53 interaction in a NR2B dependent manner, promoting oxidative stress and loss of synaptic transmission. We also evaluate a new ex vivo chronic OGD-induced ischemia model for studying the effect of oxidative stress on synaptic damage.
Collapse
Affiliation(s)
- Zhihua Zhang
- Department of Pharmacology and Toxicology, and Higuchi Bioscience Center, University of Kansas, Lawrence, KS 66045
| | - Yongfu Wang
- Department of Pharmacology and Toxicology, and Higuchi Bioscience Center, University of Kansas, Lawrence, KS 66045
| | - Shijun Yan
- Department of Pharmacology and Toxicology, and Higuchi Bioscience Center, University of Kansas, Lawrence, KS 66045
| | - Fang Du
- Department of Pharmacology and Toxicology, and Higuchi Bioscience Center, University of Kansas, Lawrence, KS 66045
| | - Shirley Shidu Yan
- Department of Pharmacology and Toxicology, and Higuchi Bioscience Center, University of Kansas, Lawrence, KS 66045
| |
Collapse
|
11
|
Kumar A, Jaggi AS, Singh N. Pharmacological investigations on possible role of Src kinases in neuroprotective mechanism of ischemic postconditioning in mice. Int J Neurosci 2014; 124:777-86. [DOI: 10.3109/00207454.2013.879869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
12
|
Qiu S, Li XY, Zhuo M. Post-translational modification of NMDA receptor GluN2B subunit and its roles in chronic pain and memory. Semin Cell Dev Biol 2011; 22:521-9. [PMID: 21704719 DOI: 10.1016/j.semcdb.2011.06.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 06/10/2011] [Accepted: 06/10/2011] [Indexed: 12/29/2022]
Abstract
N-methyl-d-aspartate receptors (NMDA receptors) play critical roles in brain functions and diseases. The expression, trafficking, synaptic location and function of different NMDA receptor subtypes are not static, but regulated dynamically in a cell-specific and synapse-specific manner during physiological and pathological conditions. In this review, we will examine recent evidence on the post-translational modulation of NMDA receptors subunit, in particular GluN2B subunit, such as phosphorylation, palmitoylation, and ubiquitination. In parallel, we will overview the roles of these modifications of GluN2B-NMDA receptor subtype in physiological functions, such as learning and memory, and pathophysiological conditions, such as chronic pain, ischemia and neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuang Qiu
- Department of Physiology, Faculty of Medicine, The Centre for the Study of Pain, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | | | | |
Collapse
|
13
|
Involvement of src-kinase activation in ischemic preconditioning induced protection of mouse brain. Life Sci 2011; 88:825-9. [PMID: 21396377 DOI: 10.1016/j.lfs.2011.02.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 02/03/2011] [Accepted: 02/24/2011] [Indexed: 10/18/2022]
Abstract
AIMS To investigate the role of src-kinase in ischemic preconditioning induced reversal of ischemia and reperfusion induced cerebral injury in mice. MAIN METHODS Bilateral carotid artery occlusion of 17min followed by reperfusion for 24h was employed to produce ischemia and reperfusion induced cerebral injury in mice. Cerebral infarct size was measured using triphenyltetrazolium chloride staining using both by volume and by weight methods differently. Memory was evaluated using elevated plus maze test. Rota rod test was employed to assess motor incoordination. KEY FINDINGS Bilateral carotid artery occlusion followed by reperfusion produced cerebral infarction and impaired memory and motor co-ordination. Three preceding episodes of bilateral carotid artery occlusion for 1min and reperfusion of 1min (ischemic preconditioning) prevented markedly ischemia-reperfusion-induced cerebral injury measured in terms of infarct size (38.5±1.3% and 38.5±2.9% mean infarct of control animals was reduced to 24.3±1.2% and 23.5±1.8% of the preconditioning groups respectively), loss of memory (72.2±3.6 mean transfer latency time of control animals was reduced to 25.6±5.2 of the preconditioning group respectively) and motor coordination (78.3±17.6s mean falling down latency time of control animals was increased to a mean value of 180.9±6.5s of the preconditioning groups respectively). SU6656 (2mg/kg, ip) and PP1 (0.1mg/kg, ip), highly selective src-kinase inhibitors, attenuated this neuroprotective effect of ischemic preconditioning. SIGNIFICANCE Therefore, neuroprotective effect of ischemic preconditioning may be due to src-kinase linked mechanism.
Collapse
|
14
|
Takagi N, Besshoh S, Morita H, Terao M, Takeo S, Tanonaka K. Metabotropic glutamate mGlu5 receptor-mediated serine phosphorylation of NMDA receptor subunit NR1 in hippocampal CA1 region after transient global ischemia in rats. Eur J Pharmacol 2010; 644:96-100. [PMID: 20667449 DOI: 10.1016/j.ejphar.2010.07.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 06/18/2010] [Accepted: 07/11/2010] [Indexed: 11/25/2022]
Abstract
Phosphorylation of the NR1 subunit of the N-methyl-d-aspartate (NMDA) receptor has been implicated in the regulation of the receptor's ion channel. The contribution of metabotropic glutamate receptors to the NMDA receptor function after brain ischemia remains to be determined. Presently we investigated the effects of an antagonist of the metabotropic glutamate mGlu5 receptor on cell death and serine phosphorylation of the NR1 subunit of the NMDA receptor in the hippocampal CA1 region after transient global ischemia and sought to explore the mechanisms involved. Phosphorylation of serine residues at 890 and 896 of NR1 was increased predominantly in the deoxycholate (DOC)-insoluble fraction after transient global ischemia in rats; and the increase in the phosphorylation of S890, but not that of S896, of NR1 in this fraction was attenuated by the mGlu5 receptor antagonist. The administration of this antagonist also reduced the increase in the amount of protein kinase C (PKC)gamma, but not that of PKCalpha, in the DOC-insoluble fraction. The results suggest that the mGlu5 receptor in the hippocampal CA1 region is involved in the phosphorylation of S890 of NR1 subunit via PKCgamma following transient ischemia. As treatment with the mGlu5 receptor antagonist reduced cell death in the hippocampal CA1 region on day 3 after the start of the reperfusion, these changes in intracellular signaling through mGlu5 receptor may be linked to the pathogenesis of cerebral ischemia.
Collapse
Affiliation(s)
- Norio Takagi
- Department of Molecular and Cellular Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| | | | | | | | | | | |
Collapse
|
15
|
Wang Y, Li Y, Dalle Lucca SL, Simovic M, Tsokos GC, Dalle Lucca JJ. Decay accelerating factor (CD55) protects neuronal cells from chemical hypoxia-induced injury. J Neuroinflammation 2010; 7:24. [PMID: 20380727 PMCID: PMC2867804 DOI: 10.1186/1742-2094-7-24] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 04/09/2010] [Indexed: 12/02/2022] Open
Abstract
Background Activated complement system is known to mediate neuroinflammation and neurodegeneration following exposure to hypoxic-ischemic insults. Therefore, inhibition of the complement activation cascade may represent a potential therapeutic strategy for the management of ischemic brain injury. Decay-accelerating factor (DAF, also known as CD55) inhibits complement activation by suppressing the function of C3/C5 convertases, thereby limiting local generation or deposition of C3a/C5a and membrane attack complex (MAC or C5b-9) production. The present study investigates the ability of DAF to protect primary cultured neuronal cells subjected to sodium cyanide (NaCN)-induced hypoxia from degeneration and apoptosis. Methods Cultured primary cortical neurons from embryonic Sprague-Dawley rats were assigned one of four groups: control, DAF treatment alone, hypoxic, or hypoxic treated with DAF. Hypoxic cultures were exposed to NaCN for 1 hour, rinsed, followed by 24 hour exposure to 200 ng/ml of recombinant human DAF in normal medium. Human DAF was used in the present study and it has been shown to effectively regulate complement activation in rats. Neuronal cell function, morphology and viability were investigated by measuring plateau depolarization potential, counting the number dendritic spines, and observing TUNEL and MTT assays. Complement C3, C3a, C3a receptor (R) production, C3a-C3aR interaction and MAC formation were assessed along with the generation of activated caspase-9, activated caspase-3, and activated Src. Results When compared to controls, hypoxic cells had fewer dendritic spines, reduced plateau depolarization accompanied by increased apoptotic activity and accumulation of MAC, as well as up-regulation of C3, C3a and C3aR, enhancement of C3a-C3aR engagement, and elevated caspase and Src activity. Treatment of hypoxic cells with 200 ng/ml of recombinant human DAF resulted in attenuation of neuronal apoptosis and exerted significant protection against neuronal dendritic spine loss and plateau depolarization reduction. Furthermore, treatment with DAF resulted in decreased accumulation of C3a, MAC, C3a-C3aR interaction, caspase-9, activated caspase-3, and pTyr416-Src (activated Src) tyrosine kinase. Conclusion DAF was found to reduce neuronal cell death and apoptosis in NaCN induced hypoxia. This effect is attributed to the ability of DAF to limit complement activation and inhibit the activity of Src and caspases 9 and 3. This study supports the inhibiting of complement as a neuroprotective strategy against CNS ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ying Wang
- Department of Cellular Injury, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | | | | | | | | | | |
Collapse
|
16
|
Bartos JA, Ulrich JD, Li H, Beazely MA, Chen Y, MacDonald JF, Hell JW. Postsynaptic clustering and activation of Pyk2 by PSD-95. J Neurosci 2010; 30:449-63. [PMID: 20071509 PMCID: PMC2822408 DOI: 10.1523/jneurosci.4992-08.2010] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2008] [Revised: 11/01/2009] [Accepted: 11/04/2009] [Indexed: 12/24/2022] Open
Abstract
The tyrosine kinase Pyk2 plays a unique role in intracellular signal transduction by linking Ca(2+) influx to tyrosine phosphorylation, but the molecular mechanism of Pyk2 activation is unknown. We report that Pyk2 oligomerization by antibodies in vitro or overexpression of PSD-95 in PC6-3 cells induces trans-autophosphorylation of Tyr402, the first step in Pyk2 activation. In neurons, Ca(2+) influx through NMDA-type glutamate receptors causes postsynaptic clustering and autophosphorylation of endogenous Pyk2 via Ca(2+)- and calmodulin-stimulated binding to PSD-95. Accordingly, Ca(2+) influx promotes oligomerization and thereby autoactivation of Pyk2 by stimulating its interaction with PSD-95. We show that this mechanism of Pyk2 activation is critical for long-term potentiation in the hippocampus CA1 region, which is thought to underlie learning and memory.
Collapse
Affiliation(s)
- Jason A. Bartos
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242-1109
| | - Jason D. Ulrich
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242-1109
| | - Hongbin Li
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada, and
| | - Michael A. Beazely
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada, and
| | - Yucui Chen
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242-1109
| | - John F. MacDonald
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada, and
- Robarts Research Institute, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | - Johannes W. Hell
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242-1109
- Department of Pharmacology, University of California, Davis, Davis, California 95616-8636
| |
Collapse
|
17
|
Tian HP, Huang BS, Zhao J, Hu XH, Guo J, Li LX. Non-receptor tyrosine kinase Src is required for ischemia-stimulated neuronal cell proliferation via Raf/ERK/CREB activation in the dentate gyrus. BMC Neurosci 2009; 10:139. [PMID: 19943942 PMCID: PMC2794287 DOI: 10.1186/1471-2202-10-139] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Accepted: 11/27/2009] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Neurogenesis in the adult mammalian hippocampus may contribute to repairing the brain after injury. However, Molecular mechanisms that regulate neuronal cell proliferation in the dentate gyrus (DG) following ischemic stroke insult are poorly understood. This study was designed to investigate the potential regulatory capacity of non-receptor tyrosine kinase Src on ischemia-stimulated cell proliferation in the adult DG and its underlying mechanism. RESULTS Src kinase activated continuously in the DG 24 h and 72 h after transient global ischemia, while SU6656, the Src kinase inhibitor significantly decreased the number of bromodeoxyuridine (BrdU) labeling-positive cells of rats 7 days after cerebral ischemia in the DG, as well as down-regulated Raf phosphorylation at Tyr(340/341) site, and its down-stream signaling molecules ERK and CREB expression followed by 24 h and 72 h of reperfusion, suggesting a role of Src kinase as an enhancer on neuronal cell proliferation in the DG via modifying the Raf/ERK/CREB cascade. This hypothesis is supported by further findings that U0126, the ERK inhibitor, induced a reduction of adult hippocampal progenitor cells in DG after cerebral ischemia and down-regulated phospho-ERK and phospho-CREB expression, but no effect was detected on the activities of Src and Raf. CONCLUSION Src kinase increase numbers of newborn neuronal cells in the DG via the activation of Raf/ERK/CREB signaling cascade after cerebral ischemia.
Collapse
Affiliation(s)
- He-Ping Tian
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Jiangsu Provincial Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China
| | - Bao-Sheng Huang
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Jiangsu Provincial Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China
| | - Jie Zhao
- Jiangsu Provincial Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Xiao-Han Hu
- Jiangsu Provincial Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Jun Guo
- Jiangsu Provincial Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing 210029, China
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Li-Xin Li
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
18
|
Marosi M, Fuzik J, Nagy D, Rákos G, Kis Z, Vécsei L, Toldi J, Ruban-Matuzani A, Teichberg VI, Farkas T. Oxaloacetate restores the long-term potentiation impaired in rat hippocampus CA1 region by 2-vessel occlusion. Eur J Pharmacol 2008; 604:51-7. [PMID: 19135048 DOI: 10.1016/j.ejphar.2008.12.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 11/20/2008] [Accepted: 12/03/2008] [Indexed: 10/21/2022]
Abstract
Various acute brain pathological conditions are characterized by the presence of elevated glutamate concentrations in the brain interstitial fluids. It has been established that a decrease in the blood glutamate level enhances the brain-to-blood efflux of glutamate, removal of which from the brain may prevent glutamate excitotoxicity and its contribution to the long-lasting neurological deficits seen in stroke. A decrease in blood glutamate level can be achieved by exploiting the glutamate-scavenging properties of the blood-resident enzyme glutamate-oxaloacetate transaminase, which transforms glutamate into 2-ketoglutarate in the presence of the glutamate co-substrate oxaloacetate. The present study had the aim of an evaluation of the effects of the blood glutamate scavenger oxaloacetate on the impaired long-term potentiation (LTP) induced in the 2-vessel occlusion ischaemic model in rat. Transient (30-min) incomplete forebrain ischaemia was produced 72 h before LTP induction. Although the short transient brain hypoperfusion did not induce histologically identifiable injuries in the CA1 region (Fluoro-Jade B, S-100 and cresyl violet), it resulted in an impaired LTP function in the hippocampal CA1 region without damaging the basal synaptic transmission between the Schaffer collaterals and the pyramidal neurons. This impairment could be fended off in a dose-dependent manner by the intravenous administration of oxaloacetate in saline (at doses between 1.5 mmol and 0.1 mumol) immediately after the transient hypoperfusion. Our results suggest that oxaloacetate-mediated blood and brain glutamate scavenging contributes to the restoration of the LTP after its impairment by brain ischaemia.
Collapse
Affiliation(s)
- Máté Marosi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Schumann J, Alexandrovich GA, Biegon A, Yaka R. Inhibition of NR2B phosphorylation restores alterations in NMDA receptor expression and improves functional recovery following traumatic brain injury in mice. J Neurotrauma 2008; 25:945-57. [PMID: 18721106 PMCID: PMC2946870 DOI: 10.1089/neu.2008.0521] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) triggers a massive glutamate efflux, hyperactivation of N-methyl-D-aspartate receptors (NMDARs) and neuronal cell death. Previously it was demonstrated that, 15 min following experimentally induced closed head injury (CHI), the density of activated NMDARs increases in the hippocampus, and decreases in the cortex at the impact site. Here we show that CHI-induced alterations in activated NMDARs correlate with changes in the expression levels of the major NMDARs subunits. In the hippocampus, the expression of NR1, NR2A, and NR2B subunits as well as the GluR1 subunit of the AMPA receptor (AMPAR) were increased, while in the cortex at the impact site, we found a decrease in the expression of these subunits. We demonstrate that CHI-induced increase in the expression of NMDAR subunits and GluR1 in the hippocampus, but not in the cortex, is associated with an increase in NR2B tyrosine phosphorylation. Furthermore, inhibition of NR2B-phosphorylation by the tyrosine kinase inhibitor PP2 restores the expression of this subunit to its normal levels. Finally, a single injection of PP2, prior to the induction of CHI, resulted in a significant improvement in long-term recovery of motor functions observed in CHI mice. These results provide a new mechanism by which acute trauma contributes to the development of secondary damage and functional deficits in the brain, and suggests a possible role for Src tyrosine kinase inhibitors as preoperative therapy for planned neurosurgical procedures.
Collapse
Affiliation(s)
- Johanna Schumann
- Department of Pharmacology, School of Pharmacy, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Anat Biegon
- Brookhaven National Laboratory, Upton, New York
| | - Rami Yaka
- Department of Pharmacology, School of Pharmacy, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
20
|
Sato Y, Tao YX, Su Q, Johns RA. Post-synaptic density-93 mediates tyrosine-phosphorylation of the N-methyl-D-aspartate receptors. Neuroscience 2008; 153:700-8. [PMID: 18423999 PMCID: PMC2696054 DOI: 10.1016/j.neuroscience.2008.03.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 02/04/2008] [Accepted: 03/02/2008] [Indexed: 11/21/2022]
Abstract
Src family protein kinases (SFKs) -mediated tyrosine-phosphorylation regulates N-methyl-D-aspartate (NMDA) receptor synaptic function. Some members of the membrane-associated guanylate kinase (MAGUK) family of proteins bind to both SFKs and NMDA receptors, but it is unclear whether the MAGUK family of proteins is required for SFKs-mediated tyrosine-phosphorylation of the NMDA receptors. Here, we showed by co-immunoprecipitation that post-synaptic density (PSD) -93, a member of the MAGUK family of proteins, interacts with the NMDA receptor subunits NR2A and NR2B as well as with Fyn, a member of the SFKs, in mouse cerebral cortex. Using a biochemical fractionation approach to isolate subcellular compartments revealed that the expression of Fyn, but not of other members of the SFKs (Lyn, Src, and Yes), was significantly decreased in synaptosomal membrane fractions derived from the cerebral cortex of PSD-93 knockout mice. Interestingly, we found that PSD-93 disruption causes reduction of tyrosine-phosphorylated NR2A and NR2B in the same fraction. Moreover, PSD-93 deletion markedly blocked the SFKs-mediated increase in tyrosine-phosphorylated NR2A and NR2B through the protein kinase C pathway after induction with 4-phorbol 12-myristate 13-acetate in cultured cortical neurons. Our findings indicate that PSD-93 appears to mediate tyrosine-phosphorylation of the NMDA receptors and synaptic localization of Fyn.
Collapse
Affiliation(s)
- Yuko Sato
- From the Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Yuan-Xiang Tao
- From the Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Qingning Su
- From the Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Roger A Johns
- From the Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
21
|
Spatiotemporal patterns of SSeCKS expression after rat spinal cord injury. Neurochem Res 2008; 33:1735-48. [PMID: 18307037 DOI: 10.1007/s11064-008-9617-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2007] [Accepted: 01/30/2008] [Indexed: 10/22/2022]
Abstract
Src suppressed C kinase substrate (SSeCKS) was identified as a PKC substrate/PKC-binding protein, which plays a role in mitogenic regulatory activity and has a function in the control of cell signaling and cytoskeletal arrangement. However its distribution and function in the central nervous system (CNS) lesion remain unclear. In this study, we mainly investigated the mRNA and protein expression and cellular localization of SSeCKS during spinal cord injury (SCI). Real-time PCR and Western blot analysis revealed that SSeCKS was present in normal whole spinal cord. It gradually increased, reached a peak at 3 days for its mRNA level and 5 days for its protein level after SCI, and then declined during the following days. In ventral horn, the expression of SSeCKS underwent a temporal pattern that was similar with the whole spinal cord in both mRNA and protein level. However, in dorsal horn, the mRNA and protein for SSeCKS expression were significantly increased at 1 day for its mRNA level and 3 days for its protein level, and then gradually declined to the baseline level, ultimately up-regulated again from 7 to 14 days. The protein expression of SSeCKS was further analysed by immunohistochemistry. The positively stained areas for SSeCKS changed with the similar pattern to that of protein expression detected by immunoblotting analysis. Double immunofluorescence staining showed that SSeCKS immunoreactivity (IR) was found in neurons, astrocytes, oligodendrocytes of spinal cord tissues within 5 mm from the lesion site. Importantly, injury-induced expression of SSeCKS was co-labeled by active caspase-3 (apoptotic marker), Tau-1 (the marker for pathological oligodendrocyte) and beta-1,4-galactosyltransferase 1 (GalT). All the results suggested that SSeCKS might play important roles in spinal cord pathophysiology and further research is needed to have a good understanding of its function and mechanism.
Collapse
|
22
|
Guo J, Wu HW, Hu G, Han X, De W, Sun YJ. Sustained activation of Src-family tyrosine kinases by ischemia: A potential mechanism mediating extracellular signal-regulated kinase cascades in hippocampal dentate gyrus. Neuroscience 2006; 143:827-36. [PMID: 17000055 DOI: 10.1016/j.neuroscience.2006.08.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2006] [Revised: 08/13/2006] [Accepted: 08/15/2006] [Indexed: 10/24/2022]
Abstract
In the present report, we investigated the association between the sustained activation of Src family tyrosine kinases (primarily Src kinase) with the biphasic phosphorylation of extracellular signal-regulated kinase (ERK) induced by ischemia in the rat hippocampal CA3/dentate gyrus subfield. Post-ischemia reperfusion resulted in the phosphorylation of ERK in a Ras-dependent manner; down-regulation of NMDA receptors or Src family protein kinases by ketamine or 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo[3,4-d] pyrimidine (PP2) potently antagonized the activation of ERK, indicating that NMDA receptors and Src family tyrosine kinases are essential for the up-regulation of ERK activity following ischemic stimuli. Additionally, an ischemia-induced association between RKIP and Raf-1 resulted in the inhibition of the ERK signaling cascade through an inhibition of Src-mediated Raf-1 phosphorylation at Tyr340/341 residues. This ischemia-induced inhibition of ERK was not associated with other downstream pathways involving Raf-1 phosphorylation at Ser 259 elicited by protein kinase B (Akt). Dissociation of Raf-1 from RKIP by 24 h reperfusion or (4S)-3-[(E)-but-2-enoyl]-4-benzyl-2-oxazolidinone (locostatin) influenced the second phase of ERK activation elicited by the Src-Raf cassette. We propose that, following ischemia, the Src family tyrosine kinases are critical for modulation of the Ras/Raf/MEK/ERK cascade, in which RKIP is involved in biphasic phosphorylation of ERK via a blockade of Src-Raf cascades.
Collapse
Affiliation(s)
- J Guo
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China.
| | | | | | | | | | | |
Collapse
|
23
|
Li Z, Zhou R, Cui S, Xie G, Cai W, Sokabe M, Chen L. Dehydroepiandrosterone sulfate prevents ischemia-induced impairment of long-term potentiation in rat hippocampal CA1 by up-regulating tyrosine phosphorylation of NMDA receptor. Neuropharmacology 2006; 51:958-66. [PMID: 16895729 DOI: 10.1016/j.neuropharm.2006.06.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 06/07/2006] [Accepted: 06/08/2006] [Indexed: 01/11/2023]
Abstract
We have reported that dehydroepiandrosterone sulfate (DHEAS) reduces the threshold for long-term potentiation (LTP) in Shaffer collateral-CA1 synapses through the amplification of Src-dependent NMDA receptor signaling. The present study is a follow-up of the above reports, aiming at evaluating the effects of DHEAS on the impaired LTP in reversible forebrain ischemic rats. Transient (20 min) incomplete forebrain ischemia led to an impaired LTP in the hippocampal CA1 region without damages to the basal synaptic transmission between the Shaffer collaterals and pyramidal neurons. Repetitive administrations of DHEAS (20 mg/kg for 3 days) from the first 3 h of reperfusion, but not acute DHEAS application (50 microM), prevent the impairment of LTP produced by ischemia. Co-administration of the specific sigma(1) receptor antagonist NE100 with DHEAS completely prevented the protective effect of DHEAS. In contrast, progesterone (PRGO) not only had no protective effect against the ischemic LTP impairment, but also attenuated the protective effect of DHEAS on the impaired LTP. Tyrosine phosphorylation of NMDA receptor subunit 2B (NR2B) significantly decreased after ischemia, whereas that of NR1 had no obvious change. Furthermore, the repetitive administration of DHEAS improved the reduction in tyrosine phosphorylation of NR2B. These findings suggest that the repetitive activation of sigma(1) receptor induced by DHEAS might prevent the ischemic LTP impairment through regulating the tyrosine phosphorylation of NR2B.
Collapse
Affiliation(s)
- Zhen Li
- Laboratory of Reproductive Medicine, Nanjing Medical University, Hanzhong Road 140, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
24
|
Huo JZ, Dykstra CM, Gurd JW. Increase in tyrosine phosphorylation of the NMDA receptor following the induction of status epilepticus. Neurosci Lett 2006; 401:266-70. [PMID: 16600505 DOI: 10.1016/j.neulet.2006.03.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2005] [Revised: 02/13/2006] [Accepted: 03/13/2006] [Indexed: 10/24/2022]
Abstract
The administration of lithium followed by pilocarpine induces status epilepticus (SE) that produces neurodegeneration and the subsequent development of spontaneous recurrent seizures. We have reported that tyrosine phosphorylation of the NMDA receptor is elevated over controls for several hours following 60 min of SE. In the current study, we assessed the temporal relationship between tyrosine phosphorylation of the NMDA receptor and the onset of SE. SE was induced using the Li/pilocarine model and phosphorylation of the NMDA receptor subunits NR2A and NR2B determined. Tyrosine phosphorylation of the NMDAR remained unchanged prior to the onset of SE and increased gradually thereafter. The onset of SE was accompanied by activation of Src-family tyrosine kinases and Pyk2 in the post-synaptic density, consistent with a role for these enzymes in SE-induced tyrosine phosphorylation. The results indicate that tyrosine phosphorylation of the NMDAR closely parallels the activation of Src-family kinases and follows, rather than precedes, the onset of SE.
Collapse
Affiliation(s)
- Jeanne Zhen Huo
- Centre for the Neurobiology of Stress, Department of Life Sciences, University of Toronto at Scarborough, Toronto, Ontario, Canada M1C 1A4
| | | | | |
Collapse
|
25
|
Goebel SM, Alvestad RM, Coultrap SJ, Browning MD. Tyrosine phosphorylation of the N-methyl-d-aspartate receptor is enhanced in synaptic membrane fractions of the adult rat hippocampus. ACTA ACUST UNITED AC 2005; 142:65-79. [PMID: 16257472 DOI: 10.1016/j.molbrainres.2005.09.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2005] [Revised: 09/01/2005] [Accepted: 09/18/2005] [Indexed: 11/30/2022]
Abstract
Hippocampal N-methyl-D-aspartate receptors (NMDARs) contribute to the expression of certain types of synaptic plasticity, such as long-term potentiation (LTP). It is well documented that tyrosine kinases increase NMDAR phosphorylation and potentiate NMDAR function. However, it is unclear how these phosphorylation changes result in enhanced NMDAR activity. We previously reported that NMDAR surface expression can be increased by LTP-inducing stimulation via tyrosine kinase-dependent mechanisms in the adult hippocampus [D.R. Grosshans, D.A. Clayton, S.J. Coultrap, M.D. Browning, Nat. Neurosci., 5 (2002) 27-33]. We therefore hypothesized that tyrosine phosphorylation of the NMDAR may enhance the trafficking of the receptor to the synaptic membrane. Here, we show that the stoichiometry of NR2A and NR2B tyrosine phosphorylation is significantly higher in synaptosomal membranes than intracellular microsomal/light membranes. Interestingly, NR2B tyrosine-1472, but not NR1 serine-896 or -897, phosphorylation is significantly higher in synaptosomal membranes than intracellular microsomal/light membranes. Furthermore, treatment of hippocampal slices with either a tyrosine phosphatase inhibitor or a tyrosine kinase inhibitor alters NMDAR tyrosine phosphorylation and produces a corresponding change in the concentration of NMDARs in the synaptosomal membrane fraction. Taken together, these data support the hypothesis that tyrosine phosphorylation may enhance NMDAR activity by increasing the number of NMDARs at the synaptic membrane.
Collapse
Affiliation(s)
- Susan M Goebel
- Neuroscience Program, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|
26
|
Abstract
Background and Purpose—
Stroke is a leading cause of disability and death in the United States, yet limited therapeutic options exist. The need for novel neuroprotective agents has spurred efforts to understand the intracellular signaling pathways that mediate cellular response to stroke. Protein kinase C (PKC) plays a central role in mediating ischemic and reperfusion damage in multiple tissues, including the brain. However, because of conflicting reports, it remains unclear whether PKC is involved in cell survival signaling, or mediates detrimental processes.
Summary of Review—
This review will examine the role of PKC activity in stroke. In particular, we will focus on more recent insights into the PKC isozyme-specific responses in neuronal preconditioning and in ischemia and reperfusion-induced stress.
Conclusion—
Examination of PKC isozyme activities during stroke demonstrates the clinical promise of PKC isozyme-specific modulators for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Rachel Bright
- Stanford University School of Medicine, Stanford, CA 94305-5174, USA
| | | |
Collapse
|
27
|
Besshoh S, Bawa D, Teves L, Wallace MC, Gurd JW. Increased phosphorylation and redistribution of NMDA receptors between synaptic lipid rafts and post-synaptic densities following transient global ischemia in the rat brain. J Neurochem 2005; 93:186-94. [PMID: 15773918 DOI: 10.1111/j.1471-4159.2004.03009.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Ischemia results in increased phosphorylation of NMDA receptors. To investigate the possible role of lipid rafts in this increase, lipid rafts and post-synaptic densities (PSDs) were isolated by the extraction of rat brain synaptosomes with Triton X-100 followed by sucrose density gradient centrifugation. Lipid rafts accounted for the majority of PSD-95, whereas SAP102 was predominantly located in PSDs. Between 50 and 60% of NMDA receptors were associated with lipid rafts. Greater than 85-90% of Src and Fyn were present in lipid rafts, whereas Pyk2 was mainly associated with PSDs. Lipid rafts and PSDs were isolated from animals subjected to 15 min of global ischemia followed by 6 h of recovery. Ischemia did not affect the yield, density, flotillin-1 or cholesterol content of lipid rafts. Following ischemia, the phosphorylation of NR1 by protein kinase C and tyrosine phosphorylation of NR2A and NR2B was increased in both lipid rafts and PSDs, with a greater increase in tyrosine phosphorylation occurring in the raft fraction. Following ischemia, NR1, NR2A and NR2B levels were elevated in PSDs and reduced in lipid rafts. The findings are consistent with a model involving close interaction between lipid rafts and PSDs and a role for lipid rafts in ischemia-induced signaling pathways.
Collapse
Affiliation(s)
- Shintaro Besshoh
- Centre for the Neurobiology of Stress, University of Toronto at Scarborough, Toronto, Canada
| | | | | | | | | |
Collapse
|
28
|
Niimura M, Moussa R, Bissoon N, Ikeda-Douglas C, Milgram NW, Gurd JW. Changes in phosphorylation of the NMDA receptor in the rat hippocampus induced by status epilepticus. J Neurochem 2005; 92:1377-85. [PMID: 15748156 DOI: 10.1111/j.1471-4159.2005.02977.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Systemic administration of pilocarpine preceded by lithium induces status epilepticus (SE) that results in neurodegeneration and may lead to the development of spontaneous recurrent seizures. We investigated the effect of Li/pilocarpine-induced SE on phosphorylation of the NMDA receptor in rat hippocampus. Phosphorylation of NR1 by PKC on Ser890 was decreased to 45% of control values immediately following 1 h of SE. During the first 3 h following the termination of SE, phosphorylation of Ser890 increased 4-fold before declining to control values by 24 h. Phosphorylation of NR1 by PKA was also depressed relative to controls immediately following SE and transiently increased above control values upon the termination of SE. SE was accompanied by a general increase in tyrosine phosphorylation of hippocampal proteins that lasted for several hours following the termination of seizures. Tyrosine phosphorylation of the NR2A and NR2B subunits of the NMDAR increased 3-4-fold over control values during SE, continued to increase during the first hour following SE and then declined to control levels by 24 h. SE resulted in the activation of Src and Pyk2 associated with the postsynaptic apparatus, suggesting a role for these enzymes in the SE-induced increase in tyrosine phosphorylation. Changes in phosphorylation of the NMDA receptor may play a role in the pathophysiological consequences of SE.
Collapse
Affiliation(s)
- M Niimura
- Centre for the Neurobiology of Stress, Department of Life Sciences, University of Toronto at Scarborough, Toronto, ON M1C 1A4, Canada
| | | | | | | | | | | |
Collapse
|
29
|
Zou X, Lin Q, Willis WD. Effect of protein kinase C blockade on phosphorylation of NR1 in dorsal horn and spinothalamic tract cells caused by intradermal capsaicin injection in rats. Brain Res 2004; 1020:95-105. [PMID: 15312791 DOI: 10.1016/j.brainres.2004.06.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2004] [Indexed: 10/26/2022]
Abstract
We have previously reported that protein kinase A (PKA) is involved in the phosphorylation of NR1 subunits of N-methyl-d-aspartate (NMDA) receptors in dorsal horn neurons after intradermal injection of capsaicin (CAP). To see if protein kinase C (PKC) also participates in the phosphorylation of NR1, we used electron microscopic techniques to determine further where the phosphorylated NR1 subunits (pNR1) are expressed in the spinothalamic tract (STT) cells and immunohistochemistry to examine whether a PKC inhibitor, chelerythrine chloride, blocks the enhanced phosphorylation of NR1 on serine 896. The pNR1 subunits were in the soma and dendrites of STT cells and in presynaptic endings. Western blots showed that pretreatment with the PKC inhibitor caused a decrease in CAP-induced phosphorylation of NR1 protein. In immunofluorescence staining, the number of pNR1-like immunoreactive neurons was significantly decreased on the side ipsilateral to the injection when chelerythrine chloride was administered intrathecally before CAP injection. In addition, when STT cells were labeled by microinjection of the retrograde tracer, fluorogold (FG), into the thalamus, we found that the proportion of p-NR1-LI STT cells was markedly reduced after PKC inhibition. Combined with our previous findings, these results strongly suggest that NR1 subunits in spinal dorsal horn neurons are phosphorylated following CAP injection, and this phosphorylation is catalyzed by PKC, as well as by PKA.
Collapse
Affiliation(s)
- Xiaoju Zou
- Department of Anatomy and Neurosciences, Marine Biomedical Institute, The University of Texas Medical Branch, Galveston, TX 77555-1069, USA
| | | | | |
Collapse
|
30
|
Affiliation(s)
- Michael W Salter
- Programme in Brain and Behaviour, The Hospital for Sick Children, University of Toronto, Ontario, Canada.
| | | |
Collapse
|