1
|
Zhang H, Chen W, Li Z, Huang Q, Wen J, Chang S, Pei H, Ma L, Li H. Huannao Yicong decoction ameliorates cognitive deficits in APP/PS1/tau triple transgenic mice by interfering with neurotoxic interaction of Aβ-tau. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116985. [PMID: 37532075 DOI: 10.1016/j.jep.2023.116985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huannao Yicong decoction (HYD) has been used in the study of AD for many years, which consists of Polygonum multiflorum Thunb., Panax ginseng C.A.Mey., Acorus gramineus Aiton, Coptis chinensis Franch., and Conioselinum acuminatum (Franch.) Lavrova. Previous studies have found that HYD could reduce β-Amyloid (Aβ) deposition and tau hyperphosphorylation which are the two critical pathological factors of AD. However, the mechanism of the neurotoxic interaction between Aβ and tau in AD remains unclear. Thus, the underlying mechanisms for HYD improving cognitive function of AD by interfering with the neurotoxic interaction between Aβ and tau remain to be explored. AIM OF THE STUDY The main objective of this study is to clarify the specific mechanisms of HYD on interfering with the neurotoxic interaction between Aβ and tau of AD both in vivo and in vitro. MATERIALS AND METHODS APP/PS1/tau triple transgenic mice were randomly divided into 4 groups, namely model group, memantine group, HYD low-dose group (HYD-L), and HYD high-dose group (HYD-H) with 28 mice in each group, while 28 C57BL/6J mice as the control group. Gavage was applied to all the mice daily for 24 weeks. SH-SY5Y model cells overexpressing Aβ and tau proteins as the intervention object in vitro experiments. Morris water maze was used to observe the learning and memory ability of APP/PS1/tau mice. Aβ deposition was detected by immunohistochemistry, and the levels of Aβ1-40 and Aβ1-42 were detected by enzyme-linked immunosorbent assay (ELISA). Neurofibrillary tangles (NFTs) were observed by silver staining and the levels of phosphorylated tau proteins were detected by Western blot. The GSK-3β and CDK-5 mRNA expression were detected by real-time polymerase chain reaction (RT-PCR). Besides, the levels of PSD95, GluR1, NR2A, and NR2B were detected by Western blot. Meanwhile, cell experiments were performed to further verify the effect of HYD on tau phosphorylation related kinases (GSK-3β, CDK-5, and PP2A), which further to clarify the mechanism of HYD intervention on the neurotoxic interaction between Aβ and tau. RESULTS HYD improved the learning and memory ability of APP/PS1/tau mice. HYD decreased the levels of Aβ1-40 and Aβ1-42 and inhibited tau hyperphosphorylation, which reduced Aβ deposition and NFTs forming. In addition, HYD inhibited the activity of kinases GSK-3β and CDK-5, and enhancing the activity of kinase PP2A. Moreover, HYD inhibited the overexpression of NR2A and NR2B, and increased the expression of GluR1 and postsynaptic density protein-95 (PSD95). CONCLUSIONS HYD can improve the cognitive deficits by interfering with the neurotoxic interaction between Aβ and tau. In addition, HYD can inhibit the overactivation of NMDARs and increase the levels of GluR1 and PSD95, which may play a role in alleviating neuronal excitotoxicity and improving synaptic function.
Collapse
Affiliation(s)
- Huiqin Zhang
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, 1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China
| | - Wenxuan Chen
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, 1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China
| | - Zehui Li
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, 1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China
| | - Qiaoyi Huang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Chaoyang District, Beijing, 100029, China
| | - Jiayu Wen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Chaoyang District, Beijing, 100029, China
| | - Surui Chang
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, 1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China
| | - Hui Pei
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, 1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China
| | - Lina Ma
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, 1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China.
| | - Hao Li
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, 1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China; Wangjing Hospital, China Academy of Chinese Medical Sciences, Hua Jia Di Jie, Chaoyang District, Beijing, 100102, China.
| |
Collapse
|
2
|
Sun Y, Chen Y, Zhan L, Zhang L, Hu J, Gao Z. The role of non-receptor protein tyrosine kinases in the excitotoxicity induced by the overactivation of NMDA receptors. Rev Neurosci 2018; 27:283-9. [PMID: 26540220 DOI: 10.1515/revneuro-2015-0037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/17/2015] [Indexed: 11/15/2022]
Abstract
Protein tyrosine phosphorylation is one of the primary modes of regulation of N-methyl-d-aspartate (NMDA) receptors. The non-receptor tyrosine kinases are one of the two types of protein tyrosine kinases that are involved in this process. The overactivation of NMDA receptors is a primary reason for neuron death following cerebral ischemia. Many studies have illustrated the important role of non-receptor tyrosine kinases in ischemia insults. This review introduces the roles of Src, Fyn, focal adhesion kinase, and proline-rich tyrosine kinase 2 in the excitotoxicity induced by the overactivation of NMDA receptors following cerebral ischemia.
Collapse
|
3
|
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: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/09/2017] [Indexed: 01/10/2023]
|
4
|
Ba M, Kong M, Ma G. Postsynaptic density protein 95-regulated NR2B tyrosine phosphorylation and interactions of Fyn with NR2B in levodopa-induced dyskinesia rat models. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 9:199-206. [PMID: 25565773 PMCID: PMC4278739 DOI: 10.2147/dddt.s75495] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Context Abnormality in interactions between N-methyl-d-aspartate (NMDA) receptor and its signaling molecules occurs in the lesioned striatum in Parkinson’s disease (PD) and levodopa-induced dyskinesia (LID). It was reported that Fyn-mediated NR2B tyrosine phosphorylation, can enhance NMDA receptor function. Postsynaptic density protein 95 (PSD-95), one of the synapse-associated proteins, regulates interactions between receptor and downstream-signaling molecules. In light of the relationship between PSD-95, NR2B, and Fyn kinases, does PSD-95 contribute to the overactivity of NMDA receptor function induced by dopaminergic treatment? To further prove the possibility, the effects of regulating the PSD-95 expression on the augmented NR2B tyrosine phosphorylation and on the interactions of Fyn and NR2B in LID rat models were evaluated. Methods In the present study, parkinsonian rat models were established by injecting 6-hydroxydopamine. Subsequently, valid PD rats were treated with levodopa (50 mg/kg/day with benserazide 12.5 mg/kg/day, twice daily) intraperitoneally for 22 days to create LID rat models. Then, the effect of pretreatment with an intrastriatal injection of the PSD-95mRNA antisense oligonucleotides (PSD-95 ASO) on the rotational response to levodopa challenge was assessed. The effects of pretreatment with an intrastriatal injection of PSD-95 ASO on the augmented NR2B tyrosine phosphorylation and interactions of Fyn with NR2B in the LID rat models were detected by immunoblotting and immunoprecipitation. Results Levodopa administration twice daily for 22 days to parkinsonian rats shortened the rotational duration and increased the peak turning responses. The altered rotational responses were attenuated by PSD-95 ASO pretreatment. Meanwhile, PSD-95 ASO pretreatment decreased the level of PSD-95 protein expression and reduced both the augmented NR2B tyrosine phosphorylation and interactions of Fyn with NR2B triggered during the levodopa administration in the lesioned striatum of PD rats. However, the protein levels of Fyn and NR2B showed no difference under the above conditions. Conclusion The data demonstrate that the inhibition of PSD-95 protein expression suppressed the interactions of Fyn with NR2B and NR2B tyrosine phosphorylation and subsequently downregulated NMDA receptor overactivation, thus providing benefit for the therapy of LID. Therefore, PSD-95 is important for overactivity of NMDA receptor function due to facilitating NR2B tyrosine phosphorylation dependent on Fyn kinase by regulating interactions of Fyn with NR2B under the pathological conditions of LID.
Collapse
Affiliation(s)
- Maowen Ba
- Department of Neurology, Yuhuangding Hospital, Yantai City, Shandong, People's Republic of China
| | - Min Kong
- Department of Neurology, Yantaishan Hospital, Yantai City, Shandong, People's Republic of China
| | - Guozhao Ma
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Shandong, People's Republic of China
| |
Collapse
|
5
|
PSD-93 deletion inhibits Fyn-mediated phosphorylation of NR2B and protects against focal cerebral ischemia. Neurobiol Dis 2014; 68:104-11. [DOI: 10.1016/j.nbd.2014.04.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 04/15/2014] [Accepted: 04/21/2014] [Indexed: 02/06/2023] Open
|
6
|
Zhao C, Du CP, Peng Y, Xu Z, Sun CC, Liu Y, Hou XY. The upregulation of NR2A-containing N-methyl-D-aspartate receptor function by tyrosine phosphorylation of postsynaptic density 95 via facilitating Src/proline-rich tyrosine kinase 2 activation. Mol Neurobiol 2014; 51:500-11. [PMID: 24981431 DOI: 10.1007/s12035-014-8796-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 06/15/2014] [Indexed: 10/25/2022]
Abstract
The activation of postsynaptic N-methyl-D-aspartate (NMDA) receptors is required for long-term potentiation (LTP) of synaptic transmission. Postsynaptic density 95 (PSD-95) serves as a scaffold protein that tethers NMDA receptor subunits, kinases, and signal molecules. Our previous study proves that PSD-95 is a substrate of Src/Fyn and identifies Y523 on PSD-95 as a principal phosphorylation site. In this paper, we try to define an involvement and molecular consequences of PSD-95 phosphorylation by Src in NMDA receptor regulation. We found that either NMDA or chemical LTP induction leads to rapid phosphorylation of PSD-95 by Src in cultured cortical neurons. The phosphorylation of Y523 on PSD-95 potentiates NR2A-containing NMDA receptor current amplitude, implying an important role of Src-mediated PSD-95 phosphorylation in NMDA receptor activation. Comparing to wild-type PSD-95, overexpression of nonphosphorylatable mutant PSD-95Y523F attenuated the NMDA-stimulated NR2A tyrosine phosphorylation that enhances electrophysiological responses of NMDA receptor channels, while did not affect the membrane localization of NR2A subunits. PSD-95Y523D, a phosphomimetic mutant of PSD-95, induced NR2A tyrosine phosphorylation even if there was no NMDA treatment. In addition, the deficiency of Y523 phosphorylation on PSD-95 impaired the facilitatory effect of PSD-95 on the activation of Src and proline-rich tyrosine kinase 2 (Pyk2) and decreased the binding of Pyk2 with PSD-95. These results indicate that PSD-95 phosphorylation by Src facilitates the integration of Pyk2 to PSD-95 signal complex, the activation of Pyk2/Src, as well as the subsequent tyrosine phosphorylation of NR2A, which ultimately results in the upregulation of NMDA receptor function and synaptic transmission.
Collapse
Affiliation(s)
- Chao Zhao
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | | | | | | | | | | | | |
Collapse
|
7
|
Beske PH, Jackson DA. NADPH oxidase mediates the oxygen-glucose deprivation/reperfusion-induced increase in the tyrosine phosphorylation of the N-methyl-D-aspartate receptor NR2A subunit in retinoic acid differentiated SH-SY5Y Cells. J Mol Signal 2012; 7:15. [PMID: 22958338 PMCID: PMC3489596 DOI: 10.1186/1750-2187-7-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 09/04/2012] [Indexed: 12/23/2022] Open
Abstract
Background Evidence exists that oxidative stress promotes the tyrosine phosphorylation of N-methyl-D-aspartate receptor (NMDAR) subunits during post-ischemic reperfusion of brain tissue. Increased tyrosine phosphorylation of NMDAR NR2A subunits has been reported to potentiate receptor function and exacerbate NMDAR-induced excitotoxicity. Though the effect of ischemia on tyrosine phosphorylation of NMDAR subunits has been well documented, the oxidative stress signaling cascades mediating the enhanced tyrosine phosphorylation of NR2A subunits remain unclear. Results We report that the reactive oxygen species (ROS) generator NADPH oxidase mediates an oxidative stress-signaling cascade involved in the increased tyrosine phosphorylation of the NR2A subunit in post-ischemic differentiated SH-SY5Y neuroblastoma cells. Inhibition of NADPH oxidase attenuated the increased tyrosine phosphorylation of the NMDAR NR2A subunit, while inhibition of ROS production from mitochondrial or xanthine oxidase sources failed to dampen the post-ischemic increase in tyrosine phosphorylation of the NR2A subunit. Additionally, inhibition of NADPH oxidase blunted the interaction of activated Src Family Kinases (SFKs) with PSD-95 induced by ischemia/reperfusion. Lastly, inhibition of NADPH oxidase also markedly reduced cell death in post-ischemic SH-SY5Y cells stimulated by NMDA. Conclusions These data indicate that NADPH oxidase has a key role in facilitating NMDAR NR2A tyrosine phosphorylation via SFK activation during post-ischemic reperfusion.
Collapse
Affiliation(s)
- Phillip H Beske
- From the Department of Biomedical and Pharmaceutical Sciences and the Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT, 59812, USA.
| | | |
Collapse
|
8
|
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: 1.1] [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
|
9
|
Hu B, Friberg H, Wieloch T. Protracted Tyrosine Phosphorylation of the Glutamate Receptor Subunit NR2 in the Rat Hippocampus Following Transient Cerebral Ischemia is Prevented by Intra-Ischemic Hypothermia. Ther Hypothermia Temp Manag 2011; 1:159-164. [PMID: 23667772 DOI: 10.1089/ther.2011.0013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Changes in the dynamic interactions of macromolecules in cell membranes appear to underlie the robust neuroprotective effect of hypothermia against selective neuronal degeneration in the CA1 region of the rat hippocampus after transient cerebral ischemia, but the detailed mechanisms are still elusive. Using the two-vessel occlusion model of transient normothermic cerebral ischemia of 15 min duration, we investigated the tyrosine phosphorylation of synaptic proteins in general and that of the NMDA receptor subunits in particular, at different times of recirculation. Specifically, the effect of intra-ischemic hypothermia (33°C), which provides neuroprotection to the CA1 region of the hippocampus, was studied. Phosphorylation of tyrosine residues on the NMDA receptor (NR) 2, but not of the NR1 or the AMPA receptor subunit 1 (GluR1) proteins, was markedly enhanced following cerebral ischemia. Protein tyrosine phosphorylation was persistently increased in the postsynaptic densities of the vulnerable CA1 region, but was transient in the CA3/dentate gyrus (DG) neurons where cell death was not evident. The phospho-tyrosine phosphatase activity decreased during reperfusion in the CA1 region but not in CA3/DG. Importantly, decreasing body temperature to 33°C during ischemia modified the dynamics of the protein tyrosine phosphorylation of NR2 in the CA1 region, which was transient and similar in time course to that seen in the CA3/DG region after normothermic ischemia. We conclude that the protracted tyrosine phosphorylation of the NR2 subunit in the hippocampus CA1 region following normothermic ischemia is attenuated by hypothermia and therefore constitutes an important target for hypothermic neuroprotection.
Collapse
Affiliation(s)
- Bingren Hu
- Laboratory for Experimental Brain Research, The Wallenberg Neuroscience Center , Lund University Hospital, Lund, Sweden . ; Shock, Trauma and Anesthesiology Research Center, Department of Anesthesiology, University of Maryland School of Medicine , Baltimore, Maryland
| | | | | |
Collapse
|
10
|
Wang WW, Hu SQ, Li C, Zhou C, Qi SH, Zhang GY. Transduced PDZ1 domain of PSD-95 decreases Src phosphorylation and increases nNOS (Ser847) phosphorylation contributing to neuroprotection after cerebral ischemia. Brain Res 2010; 1328:162-70. [DOI: 10.1016/j.brainres.2010.02.055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 02/17/2010] [Accepted: 02/17/2010] [Indexed: 11/25/2022]
|
11
|
Increased tyrosine phosphorylation of PSD-95 by Src family kinases after brain ischaemia. Biochem J 2009; 417:277-85. [PMID: 18721130 DOI: 10.1042/bj20080004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PSD (postsynaptic density)-95, a scaffold protein that tethers NMDA (N-methyl-D-aspartate) receptors to signal molecules, is implicated in pathological events resulting from excitotoxicity. The present study demonstrates that brain ischaemia and reperfusion increase the tyrosine phosphorylation of PSD-95 in the rat hippocampus. PP2, a specific inhibitor of SrcPTKs (Src family protein tyrosine kinases), prevents the ischaemia-induced increases not only in the tyrosine phosphorylation of PSD-95, but also in the interaction between PSD-95 and Src kinases. PSD-95 is phosphorylated either by purified Src/Fyn kinases in vitro or by co-expression of constitutively active Src/Fyn in COS7 cells. The results suggest that SrcPTKs are involved in PSD-95 phosphorylation. The single Tyr(523) mutation to phenylalanine (Y523F) reduces the Src/Fyn-mediated phosphorylation of PSD-95 in COS7 cells and in vitro. As shown with a rabbit polyclonal antibody against phospho-PSD-95 (Tyr(523)), Tyr(523) phosphorylation is responsible for the increased tyrosine phosphorylation of PSD-95 induced by ischaemia in the rat hippocampus. In cultured hippocampal neurons, overexpression of PSD-95 Y523F, but not PSD-95 Y533F, abolishes the facilitating effect of PSD-95 on the glutamate- or NMDA-mediated currents, implying that PSD-95 Tyr(523) phosphorylation contributes to the post-ischaemic over-activation of NMDA receptors. Thus the present study reveals an additional mechanism for the regulation of PSD-95 by tyrosine phosphorylation. This mechanism may be of pathological significance since it is associated with excitotoxicity in the ischaemic brain.
Collapse
|
12
|
The Effects of Enriched Environment on Structural Modification of Synaptic Interface and PSD-95 mRNA of Rats after Transient Focal Cerebral Ischemia. ACTA PSYCHOLOGICA SINICA 2008. [DOI: 10.3724/sp.j.1041.2008.00709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
13
|
mda-9/Syntenin promotes metastasis in human melanoma cells by activating c-Src. Proc Natl Acad Sci U S A 2008; 105:15914-9. [PMID: 18832467 DOI: 10.1073/pnas.0808171105] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The scaffold PDZ-domain containing protein mda-9/syntenin functions as a positive regulator of cancer cell progression in human melanoma and other tumors. mda-9/Syntenin regulates cell motility and invasion by altering defined biochemical and signaling pathways, including focal adhesion kinase (FAK), p38 mitogen-activated protein kinase (MAPK) and NF-kappaB, but precisely how mda-9/syntenin organizes these multiprotein signaling complexes is not well understood. Using a clinically relevant human melanoma model, we demonstrate that mda-9/syntenin physically interacts with c-Src and this communication correlates with an increase in FAK/c-Src complex formation and c-Src activation. Inhibiting mda-9/syntenin, using an adenovirus expressing antisense mda-9/syntenin or addition of c-Src siRNA, suppresses melanoma cell migration, anchorage-independent growth, and spontaneous tumor cell dissemination in vivo in a human melanoma animal metastasis model. These data are compatible with a model wherein interaction of MDA-9/syntenin with c-Src promotes the formation of an active FAK/c-Src signaling complex, leading to enhanced tumor cell invasion and metastatic spread. These provocative findings highlight mda-9/syntenin and its interacting partners as promising therapeutic targets for intervention of metastasis.
Collapse
|
14
|
Xu J, Liu Y, Zhang GY. Neuroprotection of GluR5-containing kainate receptor activation against ischemic brain injury through decreasing tyrosine phosphorylation of N-methyl-D-aspartate receptors mediated by Src kinase. J Biol Chem 2008; 283:29355-66. [PMID: 18678878 DOI: 10.1074/jbc.m800393200] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous studies indicate that cerebral ischemia breaks the dynamic balance between excitatory and inhibitory inputs. The neural excitotoxicity induced by ionotropic glutamate receptors gain the upper hand during ischemia-reperfusion. In this paper, we investigate whether GluR5 (glutamate receptor 5)-containing kainate receptor activation could lead to a neuroprotective effect against ischemic brain injury and the related mechanism. The results showed that (RS)-2-amino-3-(3-hydroxy-5-tert-butylisoxazol-4-yl) propanoic acid (ATPA), a selective GluR5 agonist, could suppress Src tyrosine phosphorylation and interactions among N-methyl-D-aspartate (NMDA) receptor subunit 2A (NR2A), postsynaptic density protein 95 (PSD-95), and Src and then decrease NMDA receptor activation through attenuating tyrosine phosphorylation of NR2A and NR2B. More importantly, ATPA had a neuroprotective effect against ischemia-reperfusion-induced neuronal cell death in vivo. However, four separate drugs were found to abolish the effects of ATPA. These were selective GluR5 antagonist NS3763; GluR5 antisense oligodeoxynucleotides; CdCl(2), a broad spectrum blocker of voltage-gated calcium channels; and bicuculline, an antagonist of gamma-aminobutyric acid A (GABA(A)) receptor. GABA(A) receptor agonist muscimol could attenuate Src activation and interactions among NR2A, PSD-95 and Src, resulting the suppression of NMDA receptor tyrosine phosphorylation. Moreover, patch clamp recording proved that the activated GABA(A) receptor could inhibit NMDA receptor-mediated whole-cell currents. Taken together, the results suggest that during ischemia-reperfusion, activated GluR5 may facilitate Ca(2+)-dependent GABA release from interneurons. The released GABA can activate postsynaptic GABA(A) receptors, which then attenuates NMDA receptor tyrosine phosphorylation through inhibiting Src activation and disassembling the signaling module NR2A-PSD-95-Src. The final result of this process is that the pyramidal neurons are rescued from hyperexcitability.
Collapse
Affiliation(s)
- Jie Xu
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bio-information, Xuzhou Medical College, Xuzhou 221002, China
| | | | | |
Collapse
|
15
|
Cai W, Zhu Y, Furuya K, Li Z, Sokabe M, Chen L. Two different molecular mechanisms underlying progesterone neuroprotection against ischemic brain damage. Neuropharmacology 2008; 55:127-38. [PMID: 18572204 DOI: 10.1016/j.neuropharm.2008.04.023] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Revised: 03/25/2008] [Accepted: 04/22/2008] [Indexed: 11/19/2022]
Abstract
Herein, we show that a single injection of P4 (4 mg/kg) at 1 h or 48 h, but not 96 h, before middle cerebral artery occlusion (MCAO) produces significant protective effects against the ischemia-induced neuronal death and the deficits in spatial cognition and LTP induction. The present study focused on the molecular mechanisms underlying the neuroprotection exerted by P4 administration at 1 h and 48 h pre-MCAO, termed acute and delayed P4-neuroprotection, respectively. Pharmacology suggested that P4-receptor (P4R) cascading to a Src-ERK1/2 signaling mediated the delayed P4-neuroprotection. To support this, it was observed by anti-phosph-ERK1/2 immunoblots that a single injection of P4 triggered a P4R-mediated persistent increase in ERK1/2 phosphorylation and their nuclear translocation for 48 h. In contrast, the acute P4-neuroprotection did not depend on the P4R-mediated Src-ERK1/2 signaling. Instead, the acute P4-administration attenuated the NMDA-induced rise in the intracellular calcium concentration ([Ca(2+)](i)) that may be a primary cause for MCAO-induced neuronal injury. This effect seemed to be exerted by an antagonism of sigma(1) receptor since the sigma(1) receptor antagonist NE100 perfectly mimicked the acute P4-neuroprotection and also attenuated the NMDA-induced [Ca(2+)](i) increase. These findings suggest that the P4 neuroprotection involves two independent processes depending on the timing of P4 administration before MCAO: an acute protection by antagonizing sigma(1) receptor to inhibit NMDAr-Ca(2+) influx and a delayed one by an activation of P4R-mediated Src-ERK signaling pathway.
Collapse
Affiliation(s)
- Weiyan Cai
- Laboratory of Reproductive Medicine, Nanjing Medical University, Hanzhong Road 140, Jiangsu, China
| | | | | | | | | | | |
Collapse
|
16
|
Jiang X, Mu D, Biran V, Faustino J, Chang S, Rincón CM, Sheldon RA, Ferriero DM. Activated Src kinases interact with theN-methyl-D-aspartate receptor after neonatal brain ischemia. Ann Neurol 2008; 63:632-41. [DOI: 10.1002/ana.21365] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
17
|
Cdk5 regulates the phosphorylation of tyrosine 1472 NR2B and the surface expression of NMDA receptors. J Neurosci 2008; 28:415-24. [PMID: 18184784 DOI: 10.1523/jneurosci.1900-07.2008] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
NMDA receptors (NMDARs) are a major class of ionotropic glutamate receptors that can undergo activity-dependent changes in surface expression. Clathrin-mediated endocytosis is a mechanism by which the surface expression of NR2B-containing NMDA receptors is regulated. The C terminus of the NMDA receptor subunit NR2B contains the internalization motif YEKL, which is the binding site for the clathrin adaptor AP-2. The tyrosine (Y1472) within the YEKL motif is phosphorylated by the Src family of kinases and this phosphorylation inhibits the binding of AP-2 and promotes surface expression of NMDA receptors. Cdk5 is a serine/threonine kinase that has been implicated in synaptic plasticity, learning, and memory. Here we demonstrate that inhibition of Cdk5 results in increased phosphorylation of Y1472 NR2B at synapses and decreased binding of NR2B to beta2-adaptin, a subunit of AP-2, thus blocking the activity-dependent endocytosis of NMDA receptors. Furthermore, we show that inhibition of Cdk5 increases the binding of Src to postsynaptic density-95 (PSD-95), and that expression of PSD-95 facilitates the phosphorylation of Y1472 NR2B by Src. Together, these results suggest a model in which inhibition of Cdk5 increases the binding of Src to PSD-95 and the phosphorylation of Y1472 NR2B by Src, which results in decreased binding of NR2B to AP-2, and NR2B/NMDAR endocytosis. This study provides a novel molecular mechanism for the regulation of the surface expression of NR2B-containing NMDA receptors and gives insight into the Cdk5-dependent regulation of synaptic plasticity.
Collapse
|
18
|
Zhang F, Li C, Wang R, Han D, Zhang QG, Zhou C, Yu HM, Zhang GY. Activation of GABA receptors attenuates neuronal apoptosis through inhibiting the tyrosine phosphorylation of NR2A by Src after cerebral ischemia and reperfusion. Neuroscience 2007; 150:938-49. [DOI: 10.1016/j.neuroscience.2007.09.070] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 09/26/2007] [Accepted: 10/05/2007] [Indexed: 10/22/2022]
|
19
|
Hou XY, Liu Y, Zhang GY. PP2, a potent inhibitor of Src family kinases, protects against hippocampal CA1 pyramidal cell death after transient global brain ischemia. Neurosci Lett 2007; 420:235-9. [PMID: 17556100 DOI: 10.1016/j.neulet.2007.03.048] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Revised: 03/20/2007] [Accepted: 03/20/2007] [Indexed: 11/30/2022]
Abstract
It has been indicated that Src family protein tyrosine kinases (SrcPTKs) potentiate N-methyl-D-aspartate (NMDA) receptor function by phosphorylating NR2A subunits and that postsynaptic density protein 95 (PSD-95) facilitates this regulation. In this paper, we define the role of SrcPTKs in delayed neuronal damage following transient brain ischemia and explore the underlying mechanisms involved in this event. Transient global brain ischemia was induced by the four-vessel occlusion method. A specific Src family kinase inhibitor PP2 (4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyramidine) and a PP2 negative control PP3 (4-amino-7-phenylpyrazolo[3,4-d]pyramidine) were infused into rat cerebroventricule 30 min before occlusion. Hematoxylin and eosine staining showed that the number of surviving pyramidal neurons in rat hippocampal CA1 subfield increased markedly in PP2-treated rats comparing to PP3-treated groups after 5 days of reperfusion following ischemia. Additionally, immunoprecipitation and immunoblot analysis revealed that preadministration of PP2, but not PP3, attenuated not only the increased tyrosine phosphorylation of NR2A but also the enhanced interactions among Src, NR2A and PSD-95 induced by ischemia/reperfusion. In conclusion, SrcPTKs promote binding of the kinases and their substrate NR2A attributed to the scaffolding effect of PSD-95 during transient brain ischemia and reperfusion, which are responsible for the elevation of NR2A tyrosine phosphorylation and consequent delayed neuronal cell death.
Collapse
Affiliation(s)
- Xiao-Yu Hou
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, 84 West Huaihai Road, Xuzhou, Jiangsu 221002, China
| | | | | |
Collapse
|
20
|
Charych EI, Akum BF, Goldberg JS, Jörnsten RJ, Rongo C, Zheng JQ, Firestein BL. Activity-independent regulation of dendrite patterning by postsynaptic density protein PSD-95. J Neurosci 2006; 26:10164-76. [PMID: 17021172 PMCID: PMC6674632 DOI: 10.1523/jneurosci.2379-06.2006] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Dendritic morphology determines many aspects of neuronal function, including action potential propagation and information processing. However, the question remains as to how distinct neuronal dendrite branching patterns are established. Here, we report that postsynaptic density-95 (PSD-95), a protein involved in dendritic spine maturation and clustering of synaptic signaling proteins, plays a novel role in regulating dendrite outgrowth and branching, independent of its synaptic functions. In immature neurons, overexpression of PSD-95 decreases the proportion of primary dendrites that undergo additional branching, resulting in a marked reduction of secondary dendrite number. Conversely, knocking down PSD-95 protein in immature neurons increases secondary dendrite number. The effect of PSD-95 is activity-independent and is antagonized by cypin, a nonsynaptic protein that regulates PSD-95 localization. Binding of cypin to PSD-95 correlates with formation of stable dendrite branches. Finally, overexpression of PSD-95 in COS-7 cells disrupts microtubule organization, indicating that PSD-95 may modulate microtubules to regulate dendritic branching. Whereas many factors have been identified which regulate dendrite number, our findings provide direct evidence that proteins primarily involved in synaptic functions can also play developmental roles in shaping how a neuron patterns its dendrite branches.
Collapse
Affiliation(s)
| | - Barbara F. Akum
- Departments of Cell Biology and Neuroscience
- Molecular Biosciences Graduate Program, Rutgers University, Piscataway, New Jersey 08854-8082, and
| | | | | | | | - James Q. Zheng
- Neuroscience and Cell Biology, The University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey 08854
| | | |
Collapse
|
21
|
Bilodeau N, Fiset A, Poirier GG, Fortier S, Gingras MC, Lavoie JN, Faure RL. Insulin-dependent phosphorylation of DPP IV in liver. Evidence for a role of compartmentalized c-Src. FEBS J 2006; 273:992-1003. [PMID: 16478473 DOI: 10.1111/j.1742-4658.2006.05125.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dipeptidyl peptidase IV (DPP IV, CD26, EC 3.4.14.5) serves as a model aimed at elucidating protein sorting signals. We identify here, by MS, several tyrosine-phosphorylated proteins in a rat liver Golgi/endosome (G/E) fraction including DPP IV. We show that a pool of DPP IV is tyrosine-phosphorylated. Maximal phosphorylation was observed after 2 min following intravenous insulin injection. DPP IV coimmunoprecipitated with the cellular tyrosine kinase Src (c-Src) with maximal association also observed after 2 min following insulin injection. DPP IV was found phosphorylated after incubation of nonsolubilized G/E membranes with [gamma-32P]ATP. The c-Src inhibitor PP2 inhibited DPP IV phosphorylation. Oriented proteolysis experiments indicate that a large pool of c-Src is protected in G/E fractions. Following injection of the protein-tyrosine phosphatase inhibitor bpV(phen), DPP IV levels markedly decreased by 40% both in plasma membrane and G/E fractions. In the fraction designated Lh, DPP IV levels decreased by 50% 15 min following insulin injection. Therefore, a pool of DPP IV is tyrosine-phosphorylated in an insulin-dependent manner. The results suggest the presence of a yet to be characterized signalling mechanism whereby DPP IV has access to c-Src-containing signalling platforms.
Collapse
Affiliation(s)
- Nicolas Bilodeau
- Pediatric Research Unit, CRCHUL/CHUQ, Faculty of Medicine, Laval University, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
22
|
Hou XY, Zhang GY, Wang DG, Guan QH, Yan JZ. Suppression of postsynaptic density protein 95 by antisense oligonucleotides diminishes postischemic pyramidal cell death in rat hippocampal CA1 subfield. Neurosci Lett 2005; 385:230-3. [PMID: 15970382 DOI: 10.1016/j.neulet.2005.05.054] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Revised: 05/04/2005] [Accepted: 05/14/2005] [Indexed: 11/27/2022]
Abstract
Our previous investigation has shown that postsynaptic density protein 95 (PSD-95) is critical for the Src family kinases-mediated tyrosine phosphorylation of N-methyl-d-aspartate receptor subunit 2A (NR2A) in the postischemic hippocampus. To clarify the roles of PSD-95 in the ischemic brain damage, histological method was performed to examine the effects of PSD-95 antisense oligonucleotides (AS) on the postischemic delayed cell death in rat hippocampus. Transient (15 min) brain ischemia was induced by the four-vessel occlusion method in Sprague-Dawley rats. Five days of reperfusion following brain ischemia (I/R5d) led to hippocampal CA1 pyramidal cell death upward of 90%. Intracerebroventricular infusion of AS (every 24 h for 3 days before ischemia) not only decreased the PSD-95 expression but also increased the number of surviving pyramidal neurons, while missense oligonucleotides (MS) had no effects. To further investigate the mechanisms underlying the neuroprotection of PSD-95 deficiency, the interaction of proline-rich tyrosine kinase 2 (Pyk2) with NR2A as well as autophosphorylation (Tyr402) of Pyk2 were detected. Immunoprecipitation and immunoblot analysis showed that preischemic treatment with AS, but not MS or vehicle, attenuated the I/R6h-induced increases in Pyk2-NR2A association and Pyk2 autophosphorylation. The protein levels of NR2A and Pyk2 had no differences under the above conditions. Our data suggest that the recruitments of ion channels and signaling molecules may be involved in the PSD-95 neurotoxicity in the postischemic hippocampus.
Collapse
Affiliation(s)
- Xiao-Yu Hou
- Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, University of Science & Technology of China, 84 West Huai-hai Road, Xuzhou, Jiangsu 221002, PR China
| | | | | | | | | |
Collapse
|
23
|
Pei DS, Sun YF, Guan QH, Hao ZB, Xu TL, Zhang GY. Postsynaptic density protein 95 antisense oligodeoxynucleotides inhibits the activation of MLK3 and JNK3 via the GluR6.PSD-95.MLK3 signaling module after transient cerebral ischemia in rat hippocampus. Neurosci Lett 2004; 367:71-5. [PMID: 15308300 DOI: 10.1016/j.neulet.2004.05.082] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2004] [Revised: 05/24/2004] [Accepted: 05/25/2004] [Indexed: 11/16/2022]
Abstract
In this study, we investigated the effect of PSD-95 antisense oligodeoxynucleotides on the phosphorylation of MLK3, JNK3 and interactions of MLK3 and PSD-95 with kainate receptor (GluR6) by immunoprecipitation and immunoblotting. Transient (15 min) brain ischemia was induced by the four-vessel occlusion in Sprague-Dawley rats. The antisense oligodeoxynucleotides of PSD-95 were administrated to the SD rats once per day for 3 days before ischemia. Our data show that the antisense oligodeoxynucleotides could inhibit phosphorylation of MLK3 and JNK3 and decrease the interactions of MLK3 and PSD-95 with GluR6. These results indicate that PSD-95 plays an important role in the formation of the GluR6.PSD-95.MLK3 signaling module and MLK3 and JNK3 activation in postischemic rat hippocampus.
Collapse
Affiliation(s)
- Dong-Sheng Pei
- Department of Neurobiology and Biophysics, School of Life Science, University of Science and Technology of China, Hefei 230027, China
| | | | | | | | | | | |
Collapse
|